1
|
Sun L, Xu L, Duan T, Xi Y, Deng Z, Luo S, Liu C, Yang C, Liu H, Sun L. CAV1 Exacerbates Renal Tubular Epithelial Cell Senescence by Suppressing CaMKK2/AMPK-Mediated Autophagy. Aging Cell 2025; 24:e14501. [PMID: 39887553 PMCID: PMC12073896 DOI: 10.1111/acel.14501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Renal proximal tubular epithelial cell (PTEC) senescence and defective autophagy contribute to kidney aging, but the mechanisms remain unclear. Caveolin-1 (CAV1), a crucial component of cell membrane caveolae, regulates autophagy and is associated with cellular senescence. However, its specific role in kidney aging is poorly understood. In this study, we generated Cav1 gene knockout mice and induced kidney aging using D-galactose (D-gal). The results showed that CAV1 expression increased in the renal cortex of the aging mice, which was accompanied by exacerbated renal interstitial fibrosis, elevated levels of senescence-associated proteins γH2AX and p16INK4a, and increased β-galactosidase activity. Moreover, autophagy and AMPK phosphorylation in PTECs were reduced. These phenotypes were partially reversed in D-gal-induced Cav1 knockout mice. Similar results were observed in D-gal-induced human proximal tubular epithelial (HK-2) cells, but these effects were blocked when AMPK activation was inhibited. Additionally, in CaMKK2 knockdown HK-2 cells, siCAV1 failed to promote AMPK phosphorylation, whereas this effect persisted when STK11 was knocked down. Besides, we examined the phosphorylation of CaMKK2 and found that siCAV1 increased its activity. Given that CaMKK2 activity is affected by intracellular Ca2+, we examined Ca2+ levels in HK-2 cells and found that D-gal treatment reduced intracellular Ca2+ concentration, but CAV1 knockdown did not alter these levels. Through GST pull-down assays, we demonstrated a direct interaction between CAV1 and CaMKK2. In conclusion, these findings suggest that CAV1 exacerbates renal tubular epithelial cell senescence by directly interacting with CaMKK2, suppressing its activity and AMPK-mediated autophagy via a Ca2+-independent pathway.
Collapse
Affiliation(s)
- Liya Sun
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood PurificationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Lujun Xu
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood PurificationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Tongyue Duan
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood PurificationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Yiyun Xi
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood PurificationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Zebin Deng
- Department of UrologyThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Shilu Luo
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood PurificationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chongbin Liu
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood PurificationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chen Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic non‐Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of NephrologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Huafeng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic non‐Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of NephrologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Lin Sun
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood PurificationThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| |
Collapse
|
2
|
Wells C, Liang Y, Pulliam TL, Lin C, Awad D, Eduful B, O’Byrne S, Hossain MA, Catta-Preta CMC, Ramos PZ, Gileadi O, Gileadi C, Couñago RM, Stork B, Langendorf CG, Nay K, Oakhill JS, Mukherjee D, Racioppi L, Means AR, York B, McDonnell DP, Scott JW, Frigo DE, Drewry DH. SGC-CAMKK2-1: A Chemical Probe for CAMKK2. Cells 2023; 12:287. [PMID: 36672221 PMCID: PMC9856672 DOI: 10.3390/cells12020287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
The serine/threonine protein kinase calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) plays critical roles in a range of biological processes. Despite its importance, only a handful of inhibitors of CAMKK2 have been disclosed. Having a selective small molecule tool to interrogate this kinase will help demonstrate that CAMKK2 inhibition can be therapeutically beneficial. Herein, we disclose SGC-CAMKK2-1, a selective chemical probe that targets CAMKK2.
Collapse
Affiliation(s)
- Carrow Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yi Liang
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas L. Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Dominik Awad
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Benjamin Eduful
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sean O’Byrne
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carolina Moura Costa Catta-Preta
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Priscila Zonzini Ramos
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Opher Gileadi
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Carina Gileadi
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Rafael M. Couñago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Brittany Stork
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Kevin Nay
- St Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia
| | | | - Debarati Mukherjee
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Luigi Racioppi
- Department of Medicine, Division of Hematological Malignancies and Cellular Therapy, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Anthony R. Means
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27705, USA
| | - John W. Scott
- St Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Daniel E. Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
3
|
Kratimenos P, Vij A, Vidva R, Koutroulis I, Delivoria-Papadopoulos M, Gallo V, Sathyanesan A. Computational analysis of cortical neuronal excitotoxicity in a large animal model of neonatal brain injury. J Neurodev Disord 2022; 14:26. [PMID: 35351004 PMCID: PMC8966144 DOI: 10.1186/s11689-022-09431-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Neonatal hypoxic brain injury is a major cause of intellectual and developmental disability. Hypoxia causes neuronal dysfunction and death in the developing cerebral cortex due to excitotoxic Ca2+-influx. In the translational piglet model of hypoxic encephalopathy, we have previously shown that hypoxia overactivates Ca2+/Calmodulin (CaM) signaling via Sarcoma (Src) kinase in cortical neurons, resulting in overexpression of proapoptotic genes. However, identifying the exact relationship between alterations in neuronal Ca2+-influx, molecular determinants of cell death, and the degree of hypoxia in a dynamic system represents a significant challenge. METHODS We used experimental and computational methods to identify molecular events critical to the onset of excitotoxicity-induced apoptosis in the cerebral cortex of newborn piglets. We used 2-3-day-old piglets (normoxic [Nx], hypoxic [Hx], and hypoxic + Src-inhibitor-treatment [Hx+PP2] groups) for biochemical analysis of ATP production, Ca2+-influx, and Ca2+/CaM-dependent protein kinase kinase 2 (CaMKK2) expression. We then used SimBiology to build a computational model of the Ca2+/CaM-Src-kinase signaling cascade, simulating Nx, Hx, and Hx+PP2 conditions. To evaluate our model, we used Sobol variance decomposition, multiparametric global sensitivity analysis, and parameter scanning. RESULTS Our model captures important molecular trends caused by hypoxia in the piglet brain. Incorporating the action of Src kinase inhibitor PP2 further validated our model and enabled predictive analysis of the effect of hypoxia on CaMKK2. We determined the impact of a feedback loop related to Src phosphorylation of NMDA receptors and activation kinetics of CaMKII. We also identified distinct modes of signaling wherein Ca2+ level alterations following Src kinase inhibition may not be a linear predictor of changes in Bax expression. Importantly, our model indicates that while pharmacological pre-treatment significantly reduces the onset of abnormal Ca2+-influx, there exists a window of intervention after hypoxia during which targeted modulation of Src-NMDAR interaction kinetics in combination with PP2 administration can reduce Ca2+-influx and Bax expression to similar levels as pre-treatment. CONCLUSIONS Our model identifies new dynamics of critical components in the Ca2+/CaM-Src signaling pathway leading to neuronal injury and provides a feasible framework for drug efficacy studies in translational models of neonatal brain injury for the prevention of intellectual and developmental disabilities.
Collapse
Affiliation(s)
- Panagiotis Kratimenos
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, 111 Michigan Avenue, Washington, DC, 20010, USA. .,Department of Pediatrics, Division of Neonatology, Children's National Hospital, Washington DC, USA. .,George Washington University School of Medicine and Health Sciences, Washington DC, USA.
| | - Abhya Vij
- George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | | | - Ioannis Koutroulis
- George Washington University School of Medicine and Health Sciences, Washington DC, USA.,Department of Pediatrics, Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA.,Center for Genetic Medicine Research, Children's National Research Institute and Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, 111 Michigan Avenue, Washington, DC, 20010, USA.,George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Aaron Sathyanesan
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, 111 Michigan Avenue, Washington, DC, 20010, USA. .,George Washington University School of Medicine and Health Sciences, Washington DC, USA.
| |
Collapse
|
4
|
Mével E, Shutter JA, Ding X, Mattingly BT, Williams JN, Li Y, Huls A, Kambrath AV, Trippel SB, Wagner D, Allen MR, O'Keefe R, Thompson WR, Burr DB, Sankar U. Systemic inhibition or global deletion of CaMKK2 protects against post-traumatic osteoarthritis. Osteoarthritis Cartilage 2022; 30:124-136. [PMID: 34506942 PMCID: PMC8712369 DOI: 10.1016/j.joca.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/17/2021] [Accepted: 09/01/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the role of Ca2+/calmodulin-dependent protein kinase 2 (CaMKK2) in post-traumatic osteoarthritis (PTOA). METHODS Destabilization of the medial meniscus (DMM) or sham surgeries were performed on 10-week-old male wild-type (WT) and Camkk2-/- mice. Half of the DMM-WT mice and all other cohorts (n = 6/group) received tri-weekly intraperitoneal (i.p.) injections of saline whereas the remaining DMM-WT mice (n = 6/group) received i.p. injections of the CaMKK2 inhibitor STO-609 (0.033 mg/kg body weight) thrice a week. Study was terminated at 8- or 12-weeks post-surgery, and knee joints processed for microcomputed tomography imaging followed by histology and immunohistochemistry. Primary articular chondrocytes were isolated from knee joints of 4-6-day-old WT and Camkk2-/- mice, and treated with 10 ng/ml interleukin-1β (IL)-1β for 24 or 48 h to investigate gene and protein expression. RESULTS CaMKK2 levels and activity became elevated in articular chondrocytes following IL-1β treatment or DMM surgery. Inhibition or absence of CaMKK2 protected against DMM-associated destruction of the cartilage, subchondral bone alterations and synovial inflammation. When challenged with IL-1β, chondrocytes lacking CaMKK2 displayed attenuated inflammation, cartilage catabolism, and resistance to suppression of matrix synthesis. IL-1β-treated CaMKK2-null chondrocytes displayed decreased IL-6 production, activation of signal transducer and activator of transcription 3 (Stat3) and matrix metalloproteinase 13 (MMP13), indicating a potential mechanism for the regulation of inflammatory responses in chondrocytes by CaMKK2. CONCLUSIONS Our findings reveal a novel function for CaMKK2 in chondrocytes and highlight the potential for its inhibition as an innovative therapeutic strategy in the prevention of PTOA.
Collapse
Affiliation(s)
- E Mével
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - J A Shutter
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - X Ding
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - B T Mattingly
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - J N Williams
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Y Li
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - A Huls
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - A V Kambrath
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - S B Trippel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - D Wagner
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Mechanical and Energy Engineering, School of Engineering and Technology, Indianapolis, IN, 46202, USA.
| | - M R Allen
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - R O'Keefe
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - W R Thompson
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indianapolis, IN, 46202, USA.
| | - D B Burr
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - U Sankar
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
5
|
Xy Ling N, Langendorf CG, Hoque A, Galic S, Loh K, Kemp BE, Gundlach AL, Oakhill JS, Scott JW. Functional analysis of an R311C variant of Ca 2+ -calmodulin-dependent protein kinase kinase-2 (CaMKK2) found as a de novo mutation in a patient with bipolar disorder. Bipolar Disord 2020; 22:841-848. [PMID: 32216002 DOI: 10.1111/bdi.12901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Loss-of-function mutations in the gene encoding the calcium-calmodulin (Ca2+ -CaM)-dependent protein kinase kinase-2 (CaMKK2) enzyme are linked to bipolar disorder. Recently, a de novo arginine to cysteine (R311C) mutation in CaMKK2 was identified from a whole exome sequencing study of bipolar patients and their unaffected parents. The aim of the present study was to determine the functional consequences of the R311C mutation on CaMKK2 activity and regulation by Ca2+ -CaM. METHODS The effects of the R311C mutation on CaMKK2 activity and Ca2+ -CaM activation were examined using a radiolabeled adenosine triphosphate (ATP) kinase assay. We performed immunoblot analysis to determine whether the R311C mutation impacts threonine-85 (T85) autophosphorylation, an activating phosphorylation site on CaMKK2 that has also been implicated in bipolar disorder. We also expressed the R311C mutant in CaMKK2 knockout HAP1 cells and used immunoblot analysis and an MTS reduction assay to study its effects on Ca2+ -dependent downstream signaling and cell viability, respectively. RESULTS The R311C mutation maps to the conserved HRD motif within the catalytic loop of CaMKK2 and caused a marked reduction in kinase activity and Ca2+ -CaM activation. The R311C mutation virtually abolished T85 autophosphorylation in response to Ca2+ -CaM and exerted a dominant-negative effect in cells as it impaired the ability of wild-type CaMKK2 to initiate downstream signaling and maintain cell viability. CONCLUSIONS The highly disruptive, loss-of-function impact of the de novo R311C mutation in human CaMKK2 provides a compelling functional rationale for being considered a potential rare monogenic cause of bipolar disorder.
Collapse
Affiliation(s)
- Naomi Xy Ling
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Christopher G Langendorf
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Ashfaqul Hoque
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Sandra Galic
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Kim Loh
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Bruce E Kemp
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Jonathan S Oakhill
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - John W Scott
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| |
Collapse
|
6
|
Effects of Single-Nucleotide Polymorphisms in Calmodulin-Dependent Protein Kinase Kinase 2 (CAMKK2): A Comprehensive Study. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:7419512. [PMID: 33082841 PMCID: PMC7559224 DOI: 10.1155/2020/7419512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 12/01/2022]
Abstract
Calmodulin-dependent protein kinase kinase 2 (CAMKK2) is a protein kinase that belongs to the serine/threonine kinase family. It phosphorylates kinases like CAMK1, CAMK2, and AMP, and this signaling cascade is involved in various biological processes including cell proliferation, apoptosis, and proliferation. Also, the CAMKK2 signaling activity is required for the healthy activity of the brain which otherwise can cause diseases like bipolar disorders and anxiety. The current study is based on in silico bioinformatics analysis that combines sequence- and structure-based predictions to mark a SNP as damaging or neutral. The combined results from sequence-based, evolutionary conservation-based, and consensus-based tools have predicted a total of 18 nsSNPs as deleterious, and these nsSNPs were further subjected to structure-based analysis. The six mutant models of V195A, V249M, R311C, F366Y, P389T, and W445C showed a higher deviation from the wildtype protein model and hence were further taken for docking studies. The molecular docking analysis has predicted that these mutations will also be disruptive to the protein-protein interactions between CAMKK2 and PRKAG1 which will create an evident reduction in the kinase activity. The current study has enlightened us that a few of the significant mutations are prime candidates in CAMKK2 which could be the fundamental cause of various bipolar and psychiatric disorders. This is the first detailed study that predicts the deleterious nsSNPs in CAMKK2 and contributes positively in providing a better understanding of disease mechanisms.
Collapse
|
7
|
Sabbir MG, Taylor CG, Zahradka P. Hypomorphic CAMKK2 in EA.hy926 endothelial cells causes abnormal transferrin trafficking, iron homeostasis and glucose metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118763. [DOI: 10.1016/j.bbamcr.2020.118763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023]
|
8
|
Langendorf CG, O'Brien MT, Ngoei KRW, McAloon LM, Dhagat U, Hoque A, Ling NXY, Dite TA, Galic S, Loh K, Parker MW, Oakhill JS, Kemp BE, Scott JW. CaMKK2 is inactivated by cAMP-PKA signaling and 14-3-3 adaptor proteins. J Biol Chem 2020; 295:16239-16250. [PMID: 32913128 DOI: 10.1074/jbc.ra120.013756] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 09/05/2020] [Indexed: 01/02/2023] Open
Abstract
The calcium-calmodulin-dependent protein kinase kinase-2 (CaMKK2) is a key regulator of cellular and whole-body energy metabolism. It is known to be activated by increases in intracellular Ca2+, but the mechanisms by which it is inactivated are less clear. CaMKK2 inhibition protects against prostate cancer, hepatocellular carcinoma, and metabolic derangements induced by a high-fat diet; therefore, elucidating the intracellular mechanisms that inactivate CaMKK2 has important therapeutic implications. Here we show that stimulation of cAMP-dependent protein kinase A (PKA) signaling in cells inactivates CaMKK2 by phosphorylation of three conserved serine residues. PKA-dependent phosphorylation of Ser495 directly impairs calcium-calmodulin activation, whereas phosphorylation of Ser100 and Ser511 mediate recruitment of 14-3-3 adaptor proteins that hold CaMKK2 in the inactivated state by preventing dephosphorylation of phospho-Ser495 We also report the crystal structure of 14-3-3ζ bound to a synthetic diphosphorylated peptide that reveals how the canonical (Ser511) and noncanonical (Ser100) 14-3-3 consensus sites on CaMKK2 cooperate to bind 14-3-3 proteins. Our findings provide detailed molecular insights into how cAMP-PKA signaling inactivates CaMKK2 and reveals a pathway to inhibit CaMKK2 with potential for treating human diseases.
Collapse
Affiliation(s)
| | - Matthew T O'Brien
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Kevin R W Ngoei
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Luke M McAloon
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Urmi Dhagat
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
| | - Ashfaqul Hoque
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Naomi X Y Ling
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Toby A Dite
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Sandra Galic
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Kim Loh
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Michael W Parker
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
| | - Jonathan S Oakhill
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Bruce E Kemp
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - John W Scott
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Australia.
| |
Collapse
|
9
|
Li B, Suutari BS, Sun SD, Luo Z, Wei C, Chenouard N, Mandelberg NJ, Zhang G, Wamsley B, Tian G, Sanchez S, You S, Huang L, Neubert TA, Fishell G, Tsien RW. Neuronal Inactivity Co-opts LTP Machinery to Drive Potassium Channel Splicing and Homeostatic Spike Widening. Cell 2020; 181:1547-1565.e15. [PMID: 32492405 DOI: 10.1016/j.cell.2020.05.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 01/28/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022]
Abstract
Homeostasis of neural firing properties is important in stabilizing neuronal circuitry, but how such plasticity might depend on alternative splicing is not known. Here we report that chronic inactivity homeostatically increases action potential duration by changing alternative splicing of BK channels; this requires nuclear export of the splicing factor Nova-2. Inactivity and Nova-2 relocation were connected by a novel synapto-nuclear signaling pathway that surprisingly invoked mechanisms akin to Hebbian plasticity: Ca2+-permeable AMPA receptor upregulation, L-type Ca2+ channel activation, enhanced spine Ca2+ transients, nuclear translocation of a CaM shuttle, and nuclear CaMKIV activation. These findings not only uncover commonalities between homeostatic and Hebbian plasticity but also connect homeostatic regulation of synaptic transmission and neuronal excitability. The signaling cascade provides a full-loop mechanism for a classic autoregulatory feedback loop proposed ∼25 years ago. Each element of the loop has been implicated previously in neuropsychiatric disease.
Collapse
Affiliation(s)
- Boxing Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China; Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA.
| | - Benjamin S Suutari
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - Simón(e) D. Sun
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - Zhengyi Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China
| | - Chuanchuan Wei
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Nicolas Chenouard
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Nataniel J Mandelberg
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Guoan Zhang
- Department of Biochemistry and Molecular Pharmacology and Skirball Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Brie Wamsley
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA 02142, USA
| | - Guoling Tian
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Sandrine Sanchez
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Sikun You
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Lianyan Huang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Thomas A Neubert
- Department of Biochemistry and Molecular Pharmacology and Skirball Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Gordon Fishell
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA 02142, USA
| | - Richard W Tsien
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
10
|
O’Byrne SN, Scott JW, Pilotte JR, Santiago ADS, Langendorf CG, Oakhill JS, Eduful BJ, Couñago RM, Wells CI, Zuercher WJ, Willson TM, Drewry DH. In Depth Analysis of Kinase Cross Screening Data to Identify CAMKK2 Inhibitory Scaffolds. Molecules 2020; 25:E325. [PMID: 31941153 PMCID: PMC7024175 DOI: 10.3390/molecules25020325] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/25/2022] Open
Abstract
The calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) activates CAMK1, CAMK4, AMPK, and AKT, leading to numerous physiological responses. The deregulation of CAMKK2 is linked to several diseases, suggesting the utility of CAMKK2 inhibitors for oncological, metabolic and inflammatory indications. In this work, we demonstrate that STO-609, frequently described as a selective inhibitor for CAMKK2, potently inhibits a significant number of other kinases. Through an analysis of literature and public databases, we have identified other potent CAMKK2 inhibitors and verified their activities in differential scanning fluorimetry and enzyme inhibition assays. These inhibitors are potential starting points for the development of selective CAMKK2 inhibitors and will lead to tools that delineate the roles of this kinase in disease biology.
Collapse
Affiliation(s)
- Sean N. O’Byrne
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - John W. Scott
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville 3052, Australia
| | - Joseph R. Pilotte
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - André da S. Santiago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas SP 13083-875, Brazil; (A.d.S.S.); (R.M.C.)
- Structural Genomics Consortium, Departamento de Genética e Evolução, Instituto de Biologia, UNICAMP, Campinas SP 13083-886, Brazil
| | - Christopher G. Langendorf
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
| | - Jonathan S. Oakhill
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia
| | - Benjamin J. Eduful
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - Rafael M. Couñago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas SP 13083-875, Brazil; (A.d.S.S.); (R.M.C.)
- Structural Genomics Consortium, Departamento de Genética e Evolução, Instituto de Biologia, UNICAMP, Campinas SP 13083-886, Brazil
| | - Carrow I. Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - William J. Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| |
Collapse
|
11
|
Yibcharoenporn C, Chusuth P, Jakakul C, Rungrotmongkol T, Chavasiri W, Muanprasat C. Discovery of a novel chalcone derivative inhibiting CFTR chloride channel via AMPK activation and its anti-diarrheal application. J Pharmacol Sci 2019; 140:273-283. [PMID: 31444000 DOI: 10.1016/j.jphs.2019.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/03/2019] [Accepted: 07/24/2019] [Indexed: 01/19/2023] Open
Abstract
Secretory diarrhea is one of the most common causes of death world-wide especially in children under 5 years old. Isoliquiritigenin (ISLQ), a plant-derived chalcone, has previously been shown to exert anti-secretory action in vitro and in vivo by inhibiting CFTR Cl- channels. However, its CFTR inhibition potency is considerably low (IC50 > 10 μM) with unknown mechanism of action. This study aimed to identify novel chalcone derivatives with improved potency and explore their mechanism of action. Screening of 27 chalcone derivatives identified CHAL-025 as the most potent chalcone analog that reversibly inhibited CFTR-mediated Cl- secretion in T84 cells with an IC50 of ∼1.5 μM. As analyzed by electrophysiological and biochemical analyses, the mechanism of CFTR inhibition by CHAL-025 is through AMP-activated protein kinase (AMPK), a negative regulator of CFTR activity. Furthermore, Western blot analyses and molecular dynamics (MD) results suggest that CHAL-025 activates AMPK by binding at the allosteric site of an upstream kinase calcium/calmodulin-dependent protein kinase kinase β (CaMKKβ). Interestingly, CHAL-025 inhibited both cholera toxin (CT) and bile acid-induced Cl- secretion in T84 cells and prevented CT-induced intestinal fluid secretion in mice. Therefore, CHAL-025 represents a promising anti-diarrheal agent that inhibits CFTR Cl- channel activity via CaMKKβ-AMPK pathways.
Collapse
Affiliation(s)
- Chamnan Yibcharoenporn
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok, 10400, Thailand
| | - Phakawat Chusuth
- Biocatalyst and Environmental Biotechnology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Phayathai, Bangkok, 10330, Thailand
| | - Chanon Jakakul
- Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Rajathevi, Bangkok, 10400, Thailand
| | - Thanyada Rungrotmongkol
- Biocatalyst and Environmental Biotechnology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Phayathai, Bangkok, 10330, Thailand; Bioinformatics and Computational Biology Program, Graduated School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Warinthorn Chavasiri
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Phayathai, Bangkok, 10330, Thailand
| | - Chatchai Muanprasat
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok, 10400, Thailand; Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok, 10400, Thailand; Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok, 10400, Thailand.
| |
Collapse
|
12
|
Expression Analysis of CYFIP1 and CAMKK2 Genes in the Blood of Epileptic and Schizophrenic Patients. J Mol Neurosci 2018; 65:336-342. [PMID: 29992499 DOI: 10.1007/s12031-018-1106-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/15/2018] [Indexed: 12/27/2022]
Abstract
Schizophrenia and epilepsy are two prevalent neurological disorders with high global burden to the society. Genome-wide studies have identified potential underlying causes for these neurological diseases. In the present case-control study, we have assessed expression of CYFIP1 and CAMKK2 genes in the blood samples of epileptic and schizophrenic patients compared with healthy subjects. A total of 180 subjects including 40 epileptic patients, 50 schizophrenic patients, and 90 healthy individuals participated in the study. Expression of the mentioned genes was measured using TaqMan real-time PCR. The results demonstrated a significant upregulation of CYFIP1 gene expression in epileptic patients (P = 0.029). CAMKK2 was downregulated in female schizophrenic patients compared with female healthy individuals (P = 0.048). These results may provide new insight into the pathogenesis of epilepsy and schizophrenia and suggest these genes as potential therapeutic targets for these neurological disorders. Future studies should evaluate these results in larger cohorts of patients.
Collapse
|
13
|
Tojkander S, Ciuba K, Lappalainen P. CaMKK2 Regulates Mechanosensitive Assembly of Contractile Actin Stress Fibers. Cell Rep 2018; 24:11-19. [DOI: 10.1016/j.celrep.2018.06.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/07/2018] [Accepted: 06/01/2018] [Indexed: 12/15/2022] Open
|
14
|
Asquith CRM, Godoi PH, Couñago RM, Laitinen T, Scott JW, Langendorf CG, Oakhill JS, Drewry DH, Zuercher WJ, Koutentis PA, Willson TM, Kalogirou AS. 1,2,6-Thiadiazinones as Novel Narrow Spectrum Calcium/Calmodulin-Dependent Protein Kinase Kinase 2 (CaMKK2) Inhibitors. Molecules 2018; 23:molecules23051221. [PMID: 29783765 PMCID: PMC6019134 DOI: 10.3390/molecules23051221] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/31/2022] Open
Abstract
We demonstrate for the first time that 4H-1,2,6-thiadiazin-4-one (TDZ) can function as a chemotype for the design of ATP-competitive kinase inhibitors. Using insights from a co-crystal structure of a 3,5-bis(arylamino)-4H-1,2,6-thiadiazin-4-one bound to calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), several analogues were identified with micromolar activity through targeted displacement of bound water molecules in the active site. Since the TDZ analogues showed reduced promiscuity compared to their 2,4-dianilinopyrimidine counter parts, they represent starting points for development of highly selective kinase inhibitors.
Collapse
Affiliation(s)
- Christopher R M Asquith
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Paulo H Godoi
- Structural Genomics Consortium, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo 13083-886, Brazil.
| | - Rafael M Couñago
- Structural Genomics Consortium, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo 13083-886, Brazil.
- Center for Molecular and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Avenida Cândido Rondon 400, P. O. Box 6010, 13083-875 Campinas, São Paulo 13083-886, Brazil.
| | - Tuomo Laitinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland.
| | - John W Scott
- St Vincent's Institute and Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia.
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia.
- The Florey Institute of Neuroscience and Mental Health, Parkville 3052, Australia.
| | - Christopher G Langendorf
- St Vincent's Institute and Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia.
| | - Jonathan S Oakhill
- St Vincent's Institute and Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia.
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia.
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - William J Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | - Timothy M Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Andreas S Kalogirou
- Department of Chemistry, University of Cyprus, P. O. Box 20537, 1678 Nicosia, Cyprus.
- Department of Life Sciences, School of Sciences, European University Cyprus, 6 Diogenis Str., Engomi, P. O. Box 22006, 1516 Nicosia, Cyprus.
| |
Collapse
|