1
|
Devasia AG, Ramasamy A, Leo CH. Current Therapeutic Landscape for Metabolic Dysfunction-Associated Steatohepatitis. Int J Mol Sci 2025; 26:1778. [PMID: 40004240 PMCID: PMC11855529 DOI: 10.3390/ijms26041778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
In recent years, "metabolic dysfunction-associated steatotic liver disease" (MASLD) has been proposed to better connect liver disease to metabolic dysfunction, which is the most common chronic liver disease worldwide. MASLD affects more than 30% of individuals globally, and it is diagnosed by the combination of hepatic steatosis and obesity, type 2 diabetes, or two metabolic risk factors. MASLD begins with the buildup of extra fat, often greater than 5%, within the liver, causing liver hepatocytes to become stressed. This can proceed to a more severe form, metabolic dysfunction-associated steatohepatitis (MASH), in 20-30% of people, where inflammation in the liver causes tissue fibrosis, which limits blood flow over time. As fibrosis worsens, MASH may lead to cirrhosis, liver failure, or even liver cancer. While the pathophysiology of MASLD is not fully known, the current "multiple-hits" concept proposes that dietary and lifestyle factors, metabolic factors, and genetic or epigenetic factors contribute to elevated oxidative stress and inflammation, causing liver fibrosis. This review article provides an overview of the pathogenesis of MASLD and evaluates existing therapies as well as pharmacological drugs that are currently being studied in clinical trials for MASLD or MASH.
Collapse
Affiliation(s)
- Arun George Devasia
- Science, Math & Technology, Singapore University of Technology & Design, Singapore 487372, Singapore;
- Genome Institute of Singapore (GIS), Agency for Science Technology and Research (A*STAR), 60 Biopolis Street, Singapore 138672, Singapore;
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore (GIS), Agency for Science Technology and Research (A*STAR), 60 Biopolis Street, Singapore 138672, Singapore;
| | - Chen Huei Leo
- Department of Biomedical Engineering, College of Design & Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
2
|
Kot A, Koszewska D, Ochman B, Świętochowska E. Clinical Potential of Misshapen/NIKs-Related Kinase (MINK) 1-A Many-Sided Element of Cell Physiology and Pathology. Curr Issues Mol Biol 2024; 46:13811-13845. [PMID: 39727954 PMCID: PMC11727420 DOI: 10.3390/cimb46120826] [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: 10/15/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Misshapen/NIKs-related kinase (MINK) 1 belongs to the mammalian germinal center kinase (GCK) family. It contains the N-terminal, conserved kinase domain, a coiled-coil region, a proline-rich region, and a GCK, C-terminal domain with the Citron-NIK-Homology (CNH) domain. The kinase is an essential component of cellular signaling pathways, which include Wnt signaling, JNK signaling, pathways engaging Ras proteins, the Hippo pathway, and STRIPAK complexes. It thus contributes to regulating the cell cycle, apoptosis, cytoskeleton organization, cell migration, embryogenesis, or tissue homeostasis. MINK1 plays an important role in immunological responses, inhibiting Th17 and Th1 cell differentiation and regulating NLRP3 inflammasome function. It may be considered a link between ROS and the immunological system, and a potential antiviral target for human enteroviruses. The kinase has been implicated in the pathogenesis of sepsis, rheumatoid arthritis, asthma, SLE, and more. It is also involved in tumorigenesis and drug resistance in cancer. Silencing MINK1 reduces cancer cell migration, suggesting potential for new therapeutic approaches. Targeting MINK1 could be a promising treatment strategy for patients insensitive to current chemotherapies, and could improve their prognosis. Moreover, MINK1 plays an important role in the nervous system and the cardiovascular system development and function. The modulation of MINK1 activity could influence the course of neurodegenerative diseases, including Alzheimer's disease. Further exploration of the activity of the kinase could also help in gaining more insight into factors involved in thrombosis or congenital heart disease. This review aims to summarize the current knowledge on MINK1, highlight its therapeutic and prognostic potential, and encourage more studies in this area.
Collapse
Affiliation(s)
| | | | | | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland; (A.K.); (D.K.); (B.O.)
| |
Collapse
|
3
|
Wunderle C, Haller L, Laager R, Bernasconi L, Neyer P, Stumpf F, Tribolet P, Stanga Z, Mueller B, Schuetz P. The Association of the Essential Amino Acids Lysine, Methionine, and Threonine with Clinical Outcomes in Patients at Nutritional Risk: Secondary Analysis of a Randomized Clinical Trial. Nutrients 2024; 16:2608. [PMID: 39203745 PMCID: PMC11357570 DOI: 10.3390/nu16162608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Lysine, methionine, and threonine are essential amino acids with vital functions for muscle and connective tissue health, metabolic balance, and the immune system. During illness, the demand for these amino acids typically increases, which puts patients at risk for deficiencies with harmful clinical consequences. In a secondary analysis of the Effect of Early Nutritional Support on Frailty, Functional Outcomes, and Recovery of Malnourished Medical Inpatients Trial (EFFORT), which compared individualized nutritional support to usual care nutrition in patients at nutritional risk, we investigated the prognostic impact of the lysine, methionine, and threonine metabolism. We had complete clinical and amino acid data in 237 patients, 58 of whom reached the primary endpoint of death at 30 days. In a model adjusted for comorbidities, sex, nutritional risk, and trial intervention, low plasma methionine levels were associated with 30-day mortality (adjusted HR 1.98 [95% CI 1.16 to 3.36], p = 0.01) and with a decline in functional status (adjusted OR 2.06 [95% CI 1.06 to 4.01], p = 0.03). The results for lysine and threonine did not show statistically significant differences regarding clinical outcomes. These findings suggest that low levels of methionine may be critical during hospitalization among patients at nutritional risk. Further studies should investigate the effect of supplementation of methionine in this patient group to improve outcomes.
Collapse
Affiliation(s)
- Carla Wunderle
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
| | - Luana Haller
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Rahel Laager
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, 3010 Bern, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland
| | - Peter Neyer
- Institute of Laboratory Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland
| | - Franziska Stumpf
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
| | - Pascal Tribolet
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Department of Health Professions, Bern University of Applied Sciences, 3008 Bern, Switzerland
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences, University of Vienna, 1030 Vienna, Austria
| | - Zeno Stanga
- Department of Diabetology, Endocrinology, Nutritional Medicine, and Metabolism, University Hospital and University of Bern, 3010 Bern, Switzerland
| | - Beat Mueller
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | - Philipp Schuetz
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
4
|
Li S, Xiong F, Zhang S, Liu J, Gao G, Xie J, Wang Y. Oligonucleotide therapies for nonalcoholic steatohepatitis. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102184. [PMID: 38665220 PMCID: PMC11044058 DOI: 10.1016/j.omtn.2024.102184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) represents a severe disease subtype of nonalcoholic fatty liver disease (NAFLD) that is thought to be highly associated with systemic metabolic abnormalities. It is characterized by a series of substantial liver damage, including hepatocellular steatosis, inflammation, and fibrosis. The end stage of NASH, in some cases, may result in cirrhosis and hepatocellular carcinoma (HCC). Nowadays a large number of investigations are actively under way to test various therapeutic strategies, including emerging oligonucleotide drugs (e.g., antisense oligonucleotide, small interfering RNA, microRNA, mimic/inhibitor RNA, and small activating RNA) that have shown high potential in treating this fatal liver disease. This article systematically reviews the pathogenesis of NASH/NAFLD, the promising druggable targets proven by current studies in chemical compounds or biological drug development, and the feasibility and limitations of oligonucleotide-based therapeutic approaches under clinical or pre-clinical studies.
Collapse
Affiliation(s)
- Sixu Li
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610066, China
| | - Feng Xiong
- Department of Cardiology, The Third People’s Hospital of Chengdu, Chengdu 610031, China
| | - Songbo Zhang
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jinghua Liu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Viral Vector Core, University of Massachusetts Chan Medical, School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Viral Vector Core, University of Massachusetts Chan Medical, School, Worcester, MA 01605, USA
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610066, China
| |
Collapse
|
5
|
Nejad FM, Mohammadabadi M, Roudbari Z, Gorji AE, Sadkowski T. Network visualization of genes involved in skeletal muscle myogenesis in livestock animals. BMC Genomics 2024; 25:294. [PMID: 38504177 PMCID: PMC10953195 DOI: 10.1186/s12864-024-10196-3] [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: 12/28/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Muscle growth post-birth relies on muscle fiber number and size. Myofibre number, metabolic and contractile capacities are established pre-birth during prenatal myogenesis. The aim of this study was to identify genes involved in skeletal muscle development in cattle, sheep, and pigs - livestock. RESULTS The cattle analysis showed significant differences in 5043 genes during the 135-280 dpc period. In sheep, 444 genes differed significantly during the 70-120 dpc period. Pigs had 905 significantly different genes for the 63-91 dpc period.The biological processes and KEGG pathway enrichment results in each species individually indicated that DEGs in cattle were significantly enriched in regulation of cell proliferation, cell division, focal adhesion, ECM-receptor interaction, and signaling pathways (PI3K-Akt, PPAR, MAPK, AMPK, Ras, Rap1); in sheep - positive regulation of fibroblast proliferation, negative regulation of endothelial cell proliferation, focal adhesion, ECM-receptor interaction, insulin resistance, and signaling pathways (PI3K-Akt, HIF-1, prolactin, Rap1, PPAR); in pigs - regulation of striated muscle tissue development, collagen fibril organization, positive regulation of insulin secretion, focal adhesion, ECM-receptor interaction, and signaling pathways (PPAR, FoxO, HIF-1, AMPK). Among the DEGs common for studied animal species, 45 common genes were identified. Based on these, a protein-protein interaction network was created and three significant modules critical for skeletal muscle myogenesis were found, with the most significant module A containing four recognized hub genes - EGFR, VEGFA, CDH1, and CAV1. Using the miRWALK and TF2DNA databases, miRNAs (bta-miR-2374 and bta-miR-744) and transcription factors (CEBPB, KLF15, RELA, ZNF143, ZBTB48, and REST) associated with hub genes were detected. Analysis of GO term and KEGG pathways showed that such processes are related to myogenesis and associated with module A: positive regulation of MAP kinase activity, vascular endothelial growth factor receptor, insulin-like growth factor binding, focal adhesion, and signaling pathways (PI3K-Akt, HIF-1, Rap1, Ras, MAPK). CONCLUSIONS The identified genes, common to the prenatal developmental period of skeletal muscle in livestock, are critical for later muscle development, including its growth by hypertrophy. They regulate valuable economic characteristics. Enhancing and breeding animals according to the recognized genes seems essential for breeders to achieve superior gains in high-quality muscle mass.
Collapse
Affiliation(s)
- Fatemeh Mohammadi Nejad
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammadreza Mohammadabadi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Zahra Roudbari
- Department of Animal Science, Faculty of Agriculture, University of Jiroft, Jiroft, Iran.
| | - Abdolvahab Ebrahimpour Gorji
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Tomasz Sadkowski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland.
| |
Collapse
|
6
|
Pagkali A, Makris A, Brofidi K, Agouridis AP, Filippatos TD. Pathophysiological Mechanisms and Clinical Associations of Non-Alcoholic Fatty Pancreas Disease. Diabetes Metab Syndr Obes 2024; 17:283-294. [PMID: 38283640 PMCID: PMC10813232 DOI: 10.2147/dmso.s397643] [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: 08/09/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Non-Alcoholic Fatty Pancreas disease (NAFPD), characterized by fat accumulation in pancreatic tissue, is an emerging clinical entity. However, the clinical associations, the underlying molecular drivers, and the pathophysiological mechanisms of NAFPD have not yet been characterized in detail. The NAFPD spectrum not only includes infiltration and accumulation of fat within and between pancreatic cells but also involves several inflammatory processes, dysregulation of physiological metabolic pathways, and hormonal defects. A deeper understanding of the underlying molecular mechanisms is key to correlate NAFPD with clinical entities including non-alcoholic fatty liver disease, metabolic syndrome, diabetes mellitus, atherosclerosis, as well as pancreatic cancer and pancreatitis. The aim of this review is to examine the pathophysiological mechanisms of NAFPD and to assess the possible causative/predictive risk factors of NAFPD-related clinical syndromes.
Collapse
Affiliation(s)
- Antonia Pagkali
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Makris
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Brofidi
- Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Aris P Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
| | | |
Collapse
|
7
|
Chen KQ, Lei GL, Ke BY, Chen L, Wang ZB, Wang SZ. STK25: a viable therapeutic target for cancer treatments? Anticancer Drugs 2023; 34:995-1001. [PMID: 36728989 DOI: 10.1097/cad.0000000000001490] [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: 02/03/2023]
Abstract
Serine/threonine protein kinase 25 (STK25) is a critical regulator of ectopic lipid storage, glucose and insulin homeostasis, fibrosis, and meta-inflammation. More and more studies have revealed a strong correlation between STK25 and human diseases. On the one hand, STK25 can affect glucose and fatty acid metabolism in normal cells or tumors. On the other hand, STK25 participates in autophagy, cell polarity, cell apoptosis, and cell migration by activating various signaling pathways. This article reviews the composition and function of STK25, the energy metabolism and potential drugs that may target STK25, and the research progress of STK25 in the occurrence and development of tumors, to provide a reference for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Ke-Qian Chen
- Department of Pharmacology, Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School
- Department of Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Guan-Lan Lei
- Department of Pharmacology, Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School
- Department of Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Bo-Yi Ke
- Department of Pharmacology, Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School
- Department of Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Lu Chen
- Department of Pharmacology, Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School
- Department of Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zong-Bao Wang
- Department of Pharmacology, Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School
- Department of Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Shu-Zhi Wang
- Department of Pharmacology, Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School
- Department of Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
8
|
Getu AA, Zhou M, Cheng SY, Tan M. The mammalian Sterile 20-like kinase 4 (MST4) signaling in tumor progression: Implications for therapy. Cancer Lett 2023; 563:216183. [PMID: 37094736 PMCID: PMC10642761 DOI: 10.1016/j.canlet.2023.216183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
Cancer is a leading cause of death in humans, with a complex and dynamic nature that makes it challenging to fully comprehend and treat. The Mammalian Sterile 20-Like Kinase 4 (MST4 or STK26) is a serine/threonine-protein kinase that plays a crucial role in cell migration and polarity in both normal and tumor cells via activation of intracellular signaling molecules and pathways. MST4 is involved in tumor cell proliferation, migration and invasion, epithelial-mesenchymal transition (EMT), survival, and cancer metastasis through modulation of downstream signaling pathways including the extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) pathways. Additionally, MST4 interacts with programmed cell death 10 (PDCD10) to promote tumor proliferation and migration. MST4 phosphorylates autophagy related 4B cysteine peptidase (ATG4B) to mediate autophagy signaling, promote tumor cell survival and proliferation, and contribute to treatment resistance. Taken together, MST4 functions as an oncogene and is a promising therapeutic target which deserves further exploration.
Collapse
Affiliation(s)
- Ayechew A Getu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Department of Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Ming Zhou
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Shi-Yuan Cheng
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Ming Tan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Institute of Biochemistry & Molecular Biology, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
9
|
Calderón-DuPont D, Torre-Villalvazo I, Díaz-Villaseñor A. Is insulin resistance tissue-dependent and substrate-specific? The role of white adipose tissue and skeletal muscle. Biochimie 2023; 204:48-68. [PMID: 36099940 DOI: 10.1016/j.biochi.2022.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/19/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023]
Abstract
Insulin resistance (IR) refers to a reduction in the ability of insulin to exert its metabolic effects in organs such as adipose tissue (AT) and skeletal muscle (SM), leading to chronic diseases such as type 2 diabetes, hepatic steatosis, and cardiovascular diseases. Obesity is the main cause of IR, however not all subjects with obesity develop clinical insulin resistance, and not all clinically insulin-resistant people have obesity. Recent evidence implies that IR onset is tissue-dependent (AT or SM) and/or substrate-specific (glucometabolic or lipometabolic). Therefore, the aims of the present review are 1) to describe the glucometabolic and lipometabolic activities of insulin in AT and SM in the maintenance of whole-body metabolic homeostasis, 2) to discuss the pathophysiology of substrate-specific IR in AT and SM, and 3) to highlight novel validated tests to assess tissue and substrate-specific IR that are easy to perform in clinical practice. In AT, glucometabolic IR reduces glucose availability for glycerol and fatty acid synthesis, thus decreasing the esterification and synthesis of signaling bioactive lipids. Lipometabolic IR in AT impairs the antilipolytic effect of insulin and lipogenesis, leading to an increase in circulating FFAs and generating lipotoxicity in peripheral tissues. In SM, glucometabolic IR reduces glucose uptake, whereas lipometabolic IR impairs mitochondrial lipid oxidation, increasing oxidative stress and inflammation, all of which lead to metabolic inflexibility. Understanding tissue-dependent and substrate-specific IR is of paramount importance for early detection before clinical manifestations and for the development of more specific treatments or direct interventions to prevent chronic life-threatening diseases.
Collapse
Affiliation(s)
- Diana Calderón-DuPont
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico; Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional en Ciencias Médicas y Nutricíon Salvador Zubirán, Mexico City, 14000, Mexico
| | - Andrea Díaz-Villaseñor
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico.
| |
Collapse
|
10
|
Zhang X, Wang BZ, Kim M, Nash TR, Liu B, Rao J, Lock R, Tamargo M, Soni RK, Belov J, Li E, Vunjak-Novakovic G, Fine B. STK25 inhibits PKA signaling by phosphorylating PRKAR1A. Cell Rep 2022; 40:111203. [PMID: 35977512 PMCID: PMC9446420 DOI: 10.1016/j.celrep.2022.111203] [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: 01/31/2022] [Revised: 06/10/2022] [Accepted: 07/21/2022] [Indexed: 11/26/2022] Open
Abstract
In the heart, protein kinase A (PKA) is critical for activating calcium handling and sarcomeric proteins in response to beta-adrenergic stimulation leading to increased myocardial contractility and performance. The catalytic activity of PKA is tightly regulated by regulatory subunits that inhibit the catalytic subunit until released by cAMP binding. Phosphorylation of type II regulatory subunits promotes PKA activation; however, the role of phosphorylation in type I regulatory subunits remain uncertain. Here, we utilize human induced pluripotent stem cell cardiomyocytes (iPSC-CMs) to identify STK25 as a kinase of the type Iα regulatory subunit PRKAR1A. Phosphorylation of PRKAR1A leads to inhibition of PKA kinase activity and increased binding to the catalytic subunit in the presence of cAMP. Stk25 knockout in mice diminishes Prkar1a phosphorylation, increases Pka activity, and augments contractile response to beta-adrenergic stimulation. Together, these data support STK25 as a negative regulator of PKA signaling through phosphorylation of PRKAR1A.
Collapse
Affiliation(s)
- Xiaokan Zhang
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Bryan Z Wang
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Michael Kim
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Trevor R Nash
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Bohao Liu
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jenny Rao
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Manuel Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - John Belov
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eric Li
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Barry Fine
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
11
|
Identification of Key Candidate Genes in Runs of Homozygosity of the Genome of Two Chicken Breeds, Associated with Cold Adaptation. BIOLOGY 2022; 11:biology11040547. [PMID: 35453746 PMCID: PMC9026094 DOI: 10.3390/biology11040547] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/22/2022] [Accepted: 03/31/2022] [Indexed: 11/20/2022]
Abstract
Simple Summary The search for genomic regions related to adaptive abilities preserved in the chicken gene pool of two breeds, which have not been under intensive selection pressure, is of great importance for breeding in the future. This study aimed to identify key candidate genes associated with the adaptation of chickens to cold environments (using the example of the Russian White breed) by using molecular genetic methods. A total of 12 key genes on breed-specific ROH (runs of homozygosity) islands were identified, which may be potential candidate genes associated with the high level of adaptability of chickens to cold environments in the early postnatal period. These genes were associated with lipid metabolism, maintaining body temperature in cold environments, non-shivering thermogenesis and muscle development and are perspectives for further research. Abstract It is well known that the chicken gene pools have high adaptive abilities, including adaptation to cold environments. This research aimed to study the genomic distribution of runs of homozygosity (ROH) in a population of Russian White (RW) chickens as a result of selection for adaptation to cold environments in the early postnatal period, to perform a structural annotation of the discovered breed-specific regions of the genome (compared to chickens of the Amroks breed) and to suggest key candidate genes associated with the adaptation of RW chickens to cold environments. Genotyping of individual samples was performed using Illumina Chicken 60K SNP BeadChip® chips. The search for homozygous regions by individual chromosomes was carried out using the PLINK 1.9 program and the detectRuns R package. Twelve key genes on breed-specific ROH islands were identified. They may be considered as potential candidate genes associated with the high adaptive ability of chickens in cold environments in the early postnatal period. Genes associated with lipid metabolism (SOCS3, NDUFA4, TXNRD2, IGFBP 1, IGFBP 3), maintaining body temperature in cold environments (ADIPOQ, GCGR, TRPM2), non-shivering thermogenesis (RYR2, CAMK2G, STK25) and muscle development (METTL21C) are perspectives for further research. This study contributes to our understanding of the mechanisms of adaptation to cold environments in chickens and provides a molecular basis for selection work.
Collapse
|
12
|
Zhou Z, Li H, Bai S, Xu Z, Jiao Y. Loss of serine/threonine protein kinase 25 in retinal ganglion cells ameliorates high glucose-elicited damage through regulation of the AKT-GSK-3β/Nrf2 pathway. Biochem Biophys Res Commun 2022; 600:87-93. [DOI: 10.1016/j.bbrc.2022.02.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/06/2022] [Accepted: 02/11/2022] [Indexed: 11/26/2022]
|
13
|
Mitrofanova A, Burke G, Merscher S, Fornoni A. New insights into renal lipid dysmetabolism in diabetic kidney disease. World J Diabetes 2021; 12:524-540. [PMID: 33995842 PMCID: PMC8107981 DOI: 10.4239/wjd.v12.i5.524] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/31/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Lipid dysmetabolism is one of the main features of diabetes mellitus and manifests by dyslipidemia as well as the ectopic accumulation of lipids in various tissues and organs, including the kidney. Research suggests that impaired cholesterol metabolism, increased lipid uptake or synthesis, increased fatty acid oxidation, lipid droplet accumulation and an imbalance in biologically active sphingolipids (such as ceramide, ceramide-1-phosphate and sphingosine-1-phosphate) contribute to the development of diabetic kidney disease (DKD). Currently, the literature suggests that both quality and quantity of lipids are associated with DKD and contribute to increased reactive oxygen species production, oxidative stress, inflammation, or cell death. Therefore, control of renal lipid dysmetabolism is a very important therapeutic goal, which needs to be archived. This article will review some of the recent advances leading to a better understanding of the mechanisms of dyslipidemia and the role of particular lipids and sphingolipids in DKD.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
| | - George Burke
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
- Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
| | - Sandra Merscher
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
| | - Alessia Fornoni
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33136, United States
| |
Collapse
|
14
|
Cansby E, Caputo M, Gao L, Kulkarni NM, Nerstedt A, Ståhlman M, Borén J, Porosk R, Soomets U, Pedrelli M, Parini P, Marschall HU, Nyström J, Howell BW, Mahlapuu M. Depletion of protein kinase STK25 ameliorates renal lipotoxicity and protects against diabetic kidney disease. JCI Insight 2020; 5:140483. [PMID: 33170807 PMCID: PMC7819747 DOI: 10.1172/jci.insight.140483] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetic kidney disease (DKD) is the most common cause of severe renal disease worldwide and the single strongest predictor of mortality in diabetes patients. Kidney steatosis has emerged as a critical trigger in the pathogenesis of DKD; however, the molecular mechanism of renal lipotoxicity remains largely unknown. Our recent studies in genetic mouse models, human cell lines, and well-characterized patient cohorts have identified serine/threonine protein kinase 25 (STK25) as a critical regulator of ectopic lipid storage in several metabolic organs prone to diabetic damage. Here, we demonstrate that overexpression of STK25 aggravates renal lipid accumulation and exacerbates structural and functional kidney injury in a mouse model of DKD. Reciprocally, inhibiting STK25 signaling in mice ameliorates diet-induced renal steatosis and alleviates the development of DKD-associated pathologies. Furthermore, we find that STK25 silencing in human kidney cells protects against lipid deposition, as well as oxidative and endoplasmic reticulum stress. Together, our results suggest that STK25 regulates a critical node governing susceptibility to renal lipotoxicity and that STK25 antagonism could mitigate DKD progression.
Collapse
Affiliation(s)
| | - Mara Caputo
- Department of Chemistry and Molecular Biology and
| | - Lei Gao
- Department of Chemistry and Molecular Biology and
| | | | | | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rando Porosk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Ursel Soomets
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | | | - Paolo Parini
- Department of Laboratory Medicine and.,Metabolism Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Brian W Howell
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | | |
Collapse
|
15
|
Basmaeil Y, Rashid MA, Khatlani T, AlShabibi M, Bahattab E, Abdullah ML, Abomaray F, Kalionis B, Massoudi S, Abumaree M. Preconditioning of Human Decidua Basalis Mesenchymal Stem/Stromal Cells with Glucose Increased Their Engraftment and Anti-diabetic Properties. Tissue Eng Regen Med 2020; 17:209-222. [PMID: 32077075 PMCID: PMC7105536 DOI: 10.1007/s13770-020-00239-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/10/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Mesenchymal stem/stromal cells (MSCs) from the decidua basalis (DBMSCs) of the human placenta have important functions that make them potential candidates for cellular therapy. Previously, we showed that DBMSC functions do not change significantly in a high oxidative stress environment, which was induced by hydrogen peroxide (H2O2) and immune cells. Here, we studied the consequences of glucose, another oxidative stress inducer, on the phenotypic and functional changes in DBMSCs. Methods: DBMSCs were exposed to a high level of glucose, and its effect on DBMSC phenotypic and functional properties was determined. DBMSC expression of oxidative stress and immune molecules after exposure to glucose were also identified. Results: Conditioning of DBMSCs with glucose improved their adhesion and invasion. Glucose also increased DBMSC expression of genes with survival, proliferation, migration, invasion, anti-inflammatory, anti-chemoattractant and antimicrobial properties. In addition, DBMSC expression of B7H4, an inhibitor of T cell proliferation was also enhanced by glucose. Interestingly, glucose modulated DBMSC expression of genes involved in insulin secretion and prevention of diabetes. Conclusion: These data show the potentially beneficial effects of glucose on DBMSC functions. Preconditioning of DBMSCs with glucose may therefore be a rational strategy for increasing their therapeutic potential by enhancing their engraftment efficiency. In addition, glucose may program DBMSCs into insulin producing cells with ability to counteract inflammation and infection associated with diabetes. However, future in vitro and in vivo studies are essential to investigate the findings of this study further. Electronic supplementary material The online version of this article (10.1007/s13770-020-00239-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yasser Basmaeil
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia.
| | - Manar Al Rashid
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia
| | - Manal AlShabibi
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Kingdom of Saudi Arabia
| | - Eman Bahattab
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Kingdom of Saudi Arabia
| | - Meshan L Abdullah
- Experimental Medicine, King Abdullah International Medical Research Center MNG-HA, Ali Al Arini, Ar Rimayah, Riyadh, 11481, Kingdom of Saudi Arabia
| | - Fawaz Abomaray
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186, Stockholm, Sweden
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre and University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Obstetrics and Gynaecology, Royal Women's Hospital, 20 Flemington Rd, Parkville, VIC, 3052, Australia
| | - Safia Massoudi
- Department of Forensic Biology, College of Forensic Sciences, Naif Arab University for Security Sciences, Khurais Rd, Ar Rimayah, Riyadh, 14812, Kingdom of Saudi Arabia
| | - Mohammad Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 1515, P.O. Box 22490, Riyadh, 11426, Kingdom of Saudi Arabia.,College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Mail Code 3124, P.O. Box 3660, Riyadh, 11481, Kingdom of Saudi Arabia
| |
Collapse
|
16
|
Abstract
One of the CCM genes, CCM3/PDCD10, binds to the protein kinase family GCKIII, which comprises MST3/STK24, SOK1/STK25, and MST4/STK26. These proteins have been shown to have the same effect as CCM3, both in endothelial cells and in animal models such as zebrafish and are most likely involved in CCM pathogenesis. We describe here an in vitro kinase assay of GCKIII proteins which can be used to study their regulation in endothelial and other cells under different circumstances.
Collapse
|
17
|
Cansby E, Magnusson E, Nuñez-Durán E, Amrutkar M, Pedrelli M, Parini P, Hoffmann J, Ståhlman M, Howell BW, Marschall HU, Borén J, Mahlapuu M. STK25 Regulates Cardiovascular Disease Progression in a Mouse Model of Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2019; 38:1723-1737. [PMID: 29930001 DOI: 10.1161/atvbaha.118.311241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective- Recent cohort studies have shown that nonalcoholic fatty liver disease (NAFLD), and especially nonalcoholic steatohepatitis (NASH), associate with atherosclerosis and cardiovascular disease, independently of conventional cardiometabolic risk factors. However, the mechanisms underlying the pathophysiological link between NAFLD/NASH and cardiovascular disease still remain unclear. Our previous studies have identified STK25 (serine/threonine protein kinase 25) as a critical determinant in ectopic lipid storage, meta-inflammation, and progression of NAFLD/NASH. The aim of this study was to assess whether STK25 is also one of the mediators in the complex molecular network controlling the cardiovascular disease risk. Approach and Results- Atherosclerosis was induced in Stk25 knockout and transgenic mice, and their wild-type littermates, by gene transfer of gain-of-function mutant of PCSK9 (proprotein convertase subtilisin/kexin type 9), which induces the downregulation of hepatic LDLR (low-density lipoprotein receptor), combined with an atherogenic western-type diet. We found that Stk25-/- mice displayed reduced atherosclerosis lesion area as well as decreased lipid accumulation, macrophage infiltration, collagen formation, and oxidative stress in aortic lesions compared with wild-type littermates, independently from alterations in dyslipidemia. Reciprocally, Stk25 transgenic mice presented aggravated plaque formation and maturation compared with wild-type littermates despite similar levels of fasting plasma cholesterol. We also found that STK25 protein was expressed in all layers of the aorta, suggesting a possible direct role in cardiovascular disease. Conclusions- This study provides the first evidence that STK25 plays a critical role in regulation of cardiovascular disease risk and suggests that pharmacological inhibition of STK25 function may provide new possibilities for prevention/treatment of atherosclerosis.
Collapse
Affiliation(s)
- Emmelie Cansby
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | - Elin Magnusson
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | - Esther Nuñez-Durán
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | - Manoj Amrutkar
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Sweden; Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, Norway (M.A.)
| | | | - Paolo Parini
- Department of Laboratory Medicine (M.P., P.P.).,Department of Medicine, Metabolism Unit (P.P.)
| | - Jenny Hoffmann
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | | | - Brian W Howell
- Karolinska Institute, Stockholm, Sweden; and Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse (B.W.H.)
| | | | - Jan Borén
- Wallenberg Laboratory (M.S., H.-U.M., J.B.)
| | - Margit Mahlapuu
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| |
Collapse
|
18
|
Pombo CM, Iglesias C, Sartages M, Zalvide JB. MST Kinases and Metabolism. Endocrinology 2019; 160:1111-1118. [PMID: 30882881 DOI: 10.1210/en.2018-00898] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022]
Abstract
Since the discovery of the mammalian sterile twenty (MST) kinase family of proteins (MST1/STK4, MST2/STK3, MST3/STK24, and SOK1/STK25), much has been done that adds to our knowledge of their structure, regulation, and function. In the last few years, a series of articles has unveiled a previous unknown relation of these kinases with metabolic regulation and the homeostasis of metabolic tissues. The aim of this review is to bring together this body of data to provide a detailed picture of the current knowledge about these proteins, metabolism, and some of the associated diseases.
Collapse
Affiliation(s)
- Celia M Pombo
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermidades Crónicas (CiMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, A Coruña, Spain
| | - Cristina Iglesias
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermidades Crónicas (CiMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, A Coruña, Spain
| | - Miriam Sartages
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermidades Crónicas (CiMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, A Coruña, Spain
| | - Juan B Zalvide
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermidades Crónicas (CiMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
19
|
Cansby E, Nuñez-Durán E, Magnusson E, Amrutkar M, Booten SL, Kulkarni NM, Svensson LT, Borén J, Marschall HU, Aghajan M, Mahlapuu M. Targeted Delivery of Stk25 Antisense Oligonucleotides to Hepatocytes Protects Mice Against Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2018; 7:597-618. [PMID: 30576769 PMCID: PMC6411916 DOI: 10.1016/j.jcmgh.2018.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are emerging as leading causes of liver disease worldwide. Currently, no specific pharmacologic therapy is available for NAFLD/NASH, which has been recognized as one of the major unmet medical needs of the 21st century. Our recent studies in genetic mouse models, human cell lines, and well-characterized patient cohorts have identified serine/threonine protein kinase (STK)25 as a critical regulator of hepatic lipid partitioning and NAFLD/NASH. Here, we studied the metabolic benefit of liver-specific STK25 inhibitors on NAFLD development and progression in a mouse model of diet-induced obesity. METHODS We developed a hepatocyte-specific triantennary N-acetylgalactosamine (GalNAc)-conjugated antisense oligonucleotide (ASO) targeting Stk25 and evaluated its effect on NAFLD features in mice after chronic exposure to dietary lipids. RESULTS We found that systemic administration of hepatocyte-targeting GalNAc-Stk25 ASO in obese mice effectively ameliorated steatosis, inflammatory infiltration, hepatic stellate cell activation, nutritional fibrosis, and hepatocellular damage in the liver compared with mice treated with GalNAc-conjugated nontargeting ASO, without any systemic toxicity or local tolerability concerns. We also observed protection against high-fat-diet-induced hepatic oxidative stress and improved mitochondrial function with Stk25 ASO treatment in mice. Moreover, GalNAc-Stk25 ASO suppressed lipogenic gene expression and acetyl-CoA carboxylase protein abundance in the liver, providing insight into the molecular mechanisms underlying repression of hepatic steatosis. CONCLUSIONS This study provides in vivo nonclinical proof-of-principle for the metabolic benefit of liver-specific inhibition of STK25 in the context of obesity and warrants future investigations to address the therapeutic potential of GalNAc-Stk25 ASO in the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Emmelie Cansby
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Esther Nuñez-Durán
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Elin Magnusson
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Manoj Amrutkar
- Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Nagaraj M. Kulkarni
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - L. Thomas Svensson
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
| | - Jan Borén
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Margit Mahlapuu
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden,Correspondence Address correspondence to: Margit Mahlapuu, PhD, Department of Chemistry & Molecular Biology, The Faculty of Science at University of Gothenburg, Medicinaregatan 9C, SE-413 90 Gothenburg, Sweden. fax: (46) 31 7862599.
| |
Collapse
|
20
|
Sütt S, Cansby E, Paul A, Amrutkar M, Nuñez-Durán E, Kulkarni NM, Ståhlman M, Borén J, Laurencikiene J, Howell BW, Enerbäck S, Mahlapuu M. STK25 regulates oxidative capacity and metabolic efficiency in adipose tissue. J Endocrinol 2018; 238:187-202. [PMID: 29794231 DOI: 10.1530/joe-18-0182] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/24/2018] [Indexed: 12/26/2022]
Abstract
Whole-body energy homeostasis at over-nutrition critically depends on how well adipose tissue remodels in response to excess calories. We recently identified serine/threonine protein kinase (STK)25 as a critical regulator of ectopic lipid storage in non-adipose tissue and systemic insulin resistance in the context of nutritional stress. Here, we investigated the role of STK25 in regulation of adipose tissue dysfunction in mice challenged with a high-fat diet. We found that overexpression of STK25 in high-fat-fed mice resulted in impaired mitochondrial function and aggravated hypertrophy, inflammatory infiltration and fibrosis in adipose depots. Reciprocally, Stk25-knockout mice displayed improved mitochondrial function and were protected against diet-induced excessive fat storage, meta-inflammation and fibrosis in brown and white adipose tissues. Furthermore, in rodent HIB-1B cell line, STK25 depletion resulted in enhanced mitochondrial activity and consequently, reduced lipid droplet size, demonstrating an autonomous action for STK25 within adipocytes. In summary, we provide the first evidence for a key function of STK25 in controlling the metabolic balance of lipid utilization vs lipid storage in brown and white adipose depots, suggesting that repression of STK25 activity offers a potential strategy for establishing healthier adipose tissue in the context of chronic exposure to dietary lipids.
Collapse
Affiliation(s)
- Silva Sütt
- Lundberg Laboratory for Diabetes ResearchDepartment of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emmelie Cansby
- Lundberg Laboratory for Diabetes ResearchDepartment of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alexandra Paul
- Department of Biology and Biological EngineeringDivision of Chemical Biology, Chalmers University of Technology, Gothenburg, Sweden
| | - Manoj Amrutkar
- Department of Hepato-Pancreato-Biliary SurgeryInstitute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Esther Nuñez-Durán
- Lundberg Laboratory for Diabetes ResearchDepartment of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Nagaraj M Kulkarni
- Lundberg Laboratory for Diabetes ResearchDepartment of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg LaboratoryInstitute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg LaboratoryInstitute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jurga Laurencikiene
- Lipid LaboratoryDepartment of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Brian W Howell
- Department of Neuroscience and PhysiologyState University of New York Upstate Medical University, Syracuse, New York, USA
| | - Sven Enerbäck
- Department of Medical and Clinical GeneticsInstitute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Margit Mahlapuu
- Lundberg Laboratory for Diabetes ResearchDepartment of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
21
|
Wu F, Gao P, Wu W, Wang Z, Yang J, Di J, Jiang B, Su X. STK25-induced inhibition of aerobic glycolysis via GOLPH3-mTOR pathway suppresses cell proliferation in colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:144. [PMID: 29996891 PMCID: PMC6042396 DOI: 10.1186/s13046-018-0808-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/07/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Serine/threonine protein kinase 25 (STK25) is critical in regulating whole-body glucose and insulin homeostasis and the accumulation of ectopic lipids. The Warburg effect, also known as aerobic glycolysis, is an essential metabolic characteristic of cancer cells. However, the effects of STK25 on aerobic glycolysis of cancer cells remain unexplored. The aim of this study is to investigate the role of STK25 in colorectal cancer (CRC) and to elucidate the underlying mechanisms. METHODS The influences of STK25 on the cell proliferation were evaluated by MTT and colony formation assays. The roles of STK25 in aerobic glycolysis were determined by glucose uptake and lactate production assays. The interaction between STK25 and GOLPH3 was detected by co-immunoprecipitation, GST pull-down, and His-tag pull-down assays. Western blot was used to measure the expression of glycolytic genes, and the status of kinases in mTOR pathway. Moreover, a xenograft mouse model was used to investigate the effects of STK25 in vivo. The prognostic significance of STK25 was analyzed using public CRC datasets by a log-rank test. RESULTS STK25 suppressed proliferation, glycolysis and glycolytic gene expression in CRC cells. STK25 interacted with GOLPH3 and mediated glycolysis through GOLPH3-regulated mTOR signaling. Consistent with these observations, silencing of STK25 promoted tumor growth and glycolytic gene expression in an in vivo xenograft mouse model. Moreover, high levels of STK25 correlated with favorable prognosis in patients with CRC. CONCLUSIONS Our results demonstrated that STK25 negatively regulates the proliferation and glycolysis via GOLPH3-dependent mTOR signaling. Accordingly, STK25 could be a potential therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Fan Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.,Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Pin Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Wei Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Zaozao Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jie Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jiabo Di
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Beihai Jiang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Xiangqian Su
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
22
|
Iglesias C, Floridia E, Sartages M, Porteiro B, Fraile M, Guerrero A, Santos D, Cuñarro J, Tovar S, Nogueiras R, Pombo CM, Zalvide J. The MST3/STK24 kinase mediates impaired fasting blood glucose after a high-fat diet. Diabetologia 2017; 60:2453-2462. [PMID: 28956081 DOI: 10.1007/s00125-017-4433-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/28/2017] [Indexed: 01/15/2023]
Abstract
AIMS/HYPOTHESIS The identification of mediators in the pathogenesis of type 2 diabetes mellitus is essential for the full understanding of this disease. Protein kinases are especially important because of their potential as pharmacological targets. The goal of this study was to investigate whether mammalian sterile-20 3 (MST3/STK24), a stress-regulated kinase, is involved in metabolic alterations in obesity. METHODS Glucose regulation of Mst3 (also known as Stk24)-knockout mice was analysed both in 129;C57 mixed background mice and in C57/BL6J mice fed normally or with a high-fat diet (HFD). This work was complemented with an analysis of the insulin signalling pathway in cultured human liver cells made deficient in MST3 using RNA interference. RESULTS MST3 is phosphorylated in the livers of mice subject to an obesity-promoting HFD, and its deficiency lowers the hyperglycaemia, hyperinsulinaemia and insulin resistance that the animals develop with this diet, an effect that is seen even without complete inactivation of the kinase. Lack of MST3 results in activation of the insulin signalling pathway downstream of IRS1, in both cultured liver cells and the liver of animals after HFD. This effect increases the inhibition of forkhead box (FOX)O1, with subsequent downregulation of the expression of gluconeogenic enzymes. CONCLUSIONS/INTERPRETATION MST3 inhibits the insulin signalling pathway and is important in the development of insulin resistance and impaired blood glucose levels after an HFD.
Collapse
Affiliation(s)
- Cristina Iglesias
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Ebel Floridia
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Miriam Sartages
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Begoña Porteiro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - María Fraile
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Ana Guerrero
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, London, UK
| | - Diana Santos
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
| | - Juan Cuñarro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Sulay Tovar
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Rubén Nogueiras
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Celia M Pombo
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain.
| | - Juan Zalvide
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular e Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Avda de Barcelona s/n, 15706 A, Santiago de Compostela, Coruña, Spain.
| |
Collapse
|
23
|
Nuñez-Durán E, Aghajan M, Amrutkar M, Sütt S, Cansby E, Booten SL, Watt A, Ståhlman M, Stefan N, Häring HU, Staiger H, Borén J, Marschall HU, Mahlapuu M. Serine/threonine protein kinase 25 antisense oligonucleotide treatment reverses glucose intolerance, insulin resistance, and nonalcoholic fatty liver disease in mice. Hepatol Commun 2017; 2:69-83. [PMID: 29404514 PMCID: PMC5776874 DOI: 10.1002/hep4.1128] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/16/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) contributes to the pathogenesis of type 2 diabetes and cardiovascular disease, and patients with nonalcoholic steatohepatitis (NASH) are also at risk of developing cirrhosis, liver failure, and hepatocellular carcinoma. To date, no specific therapy exists for NAFLD/NASH, which has been recognized as one of the major unmet medical needs of the twenty‐first century. We recently identified serine/threonine protein kinase (STK)25 as a critical regulator of energy homeostasis and NAFLD progression. Here, we investigated the effect of antisense oligonucleotides (ASOs) targeting Stk25 on the metabolic and molecular phenotype of mice after chronic exposure to dietary lipids. We found that Stk25 ASOs efficiently reversed high‐fat diet‐induced systemic hyperglycemia and hyperinsulinemia, improved whole‐body glucose tolerance and insulin sensitivity, and ameliorated liver steatosis, inflammatory infiltration, apoptosis, hepatic stellate cell activation, and nutritional fibrosis in obese mice. Moreover, Stk25 ASOs suppressed the abundance of liver acetyl‐coenzyme A carboxylase (ACC) protein, a key regulator of both lipid oxidation and synthesis, revealing the likely mechanism underlying repression of hepatic fat accumulation by ASO treatment. We also found that STK25 protein levels correlate significantly and positively with NASH development in human liver biopsies, and several common nonlinked single‐nucleotide polymorphisms in the human STK25 gene are associated with altered liver fat, supporting a critical role of STK25 in the pathogenesis of NAFLD in humans. Conclusion: Preclinical validation for the metabolic benefit of pharmacologically inhibiting STK25 in the context of obesity is provided. Therapeutic intervention aimed at reducing STK25 function may provide a new strategy for the treatment of patients with NAFLD, type 2 diabetes, and related complex metabolic diseases. (Hepatology Communications 2018;2:69–83)
Collapse
Affiliation(s)
- Esther Nuñez-Durán
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | | | - Manoj Amrutkar
- Department of Gastrointestinal and Children Surgery University of Oslo Norway
| | - Silva Sütt
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | - Emmelie Cansby
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | | | | | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich University of Tübingen Tübingen Germany.,German Center for Diabetes Research Tübingen Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich University of Tübingen Tübingen Germany.,German Center for Diabetes Research Tübingen Germany
| | - Harald Staiger
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich University of Tübingen Tübingen Germany.,German Center for Diabetes Research Tübingen Germany.,Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry Eberhard Karls University Tübingen Tübingen Germany
| | - Jan Borén
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| | - Margit Mahlapuu
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Sahlgrenska University Hospital Gothenburg Sweden
| |
Collapse
|
24
|
Pravenec M, Saba LM, Zídek V, Landa V, Mlejnek P, Šilhavý J, Šimáková M, Strnad H, Trnovská J, Škop V, Hüttl M, Marková I, Oliyarnyk O, Malínská H, Kazdová L, Smith H, Tabakoff B. Systems genetic analysis of brown adipose tissue function. Physiol Genomics 2017; 50:52-66. [PMID: 29127223 DOI: 10.1152/physiolgenomics.00091.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Brown adipose tissue (BAT) has been suggested to play an important role in lipid and glucose metabolism in rodents and possibly also in humans. In the current study, we used genetic and correlation analyses in the BXH/HXB recombinant inbred (RI) strains, derived from Brown Norway (BN) and spontaneously hypertensive rats (SHR), to identify genetic determinants of BAT function. Linkage analyses revealed a quantitative trait locus (QTL) associated with interscapular BAT mass on chromosome 4 and two closely linked QTLs associated with glucose oxidation and glucose incorporation into BAT lipids on chromosome 2. Using weighted gene coexpression network analysis (WGCNA) we identified 1,147 gene coexpression modules in the BAT from BXH/HXB rats and mapped their module eigengene QTLs. Through an unsupervised analysis, we identified modules related to BAT relative mass and function. The Coral4.1 coexpression module is associated with BAT relative mass (includes Cd36 highly connected gene), and the Darkseagreen coexpression module is associated with glucose incorporation into BAT lipids (includes Hiat1, Fmo5, and Sort1 highly connected transcripts). Because multiple statistical criteria were used to identify candidate modules, significance thresholds for individual tests were not adjusted for multiple comparisons across modules. In summary, a systems genetic analysis using genomic and quantitative transcriptomic and physiological information has produced confirmation of several known genetic factors and significant insight into novel genetic components functioning in BAT and possibly contributing to traits characteristic of the metabolic syndrome.
Collapse
Affiliation(s)
- Michal Pravenec
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Laura M Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Václav Zídek
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Vladimír Landa
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Petr Mlejnek
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Jan Šilhavý
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Miroslava Šimáková
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Hynek Strnad
- Institute of Molecular Genetics of the Czech Academy of Sciences , Prague , Czech Republic
| | - Jaroslava Trnovská
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vojtěch Škop
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martina Hüttl
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Irena Marková
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Olena Oliyarnyk
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Hana Malínská
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ludmila Kazdová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Harry Smith
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, Colorado.,Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| |
Collapse
|
25
|
Nuñez-Durán E, Chanclón B, Sütt S, Real J, Marschall HU, Wernstedt Asterholm I, Cansby E, Mahlapuu M. Protein kinase STK25 aggravates the severity of non-alcoholic fatty pancreas disease in mice. J Endocrinol 2017; 234:15-27. [PMID: 28442507 PMCID: PMC5510597 DOI: 10.1530/joe-17-0018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/25/2017] [Indexed: 12/18/2022]
Abstract
Characterising the molecular networks that negatively regulate pancreatic β-cell function is essential for understanding the underlying pathogenesis and developing new treatment strategies for type 2 diabetes. We recently identified serine/threonine protein kinase 25 (STK25) as a critical regulator of ectopic fat storage, meta-inflammation, and fibrosis in liver and skeletal muscle. Here, we assessed the role of STK25 in control of progression of non-alcoholic fatty pancreas disease in the context of chronic exposure to dietary lipids in mice. We found that overexpression of STK25 in high-fat-fed transgenic mice aggravated diet-induced lipid storage in the pancreas compared with that of wild-type controls, which was accompanied by exacerbated pancreatic inflammatory cell infiltration, stellate cell activation, fibrosis and apoptosis. Pancreas of Stk25 transgenic mice also displayed a marked decrease in islet β/α-cell ratio and alteration in the islet architecture with an increased presence of α-cells within the islet core, whereas islet size remained similar between genotypes. After a continued challenge with a high-fat diet, lower levels of fasting plasma insulin and C-peptide, and higher levels of plasma leptin, were detected in Stk25 transgenic vs wild-type mice. Furthermore, the glucose-stimulated insulin secretion was impaired in high-fat-fed Stk25 transgenic mice during glucose tolerance test, in spite of higher net change in blood glucose concentrations compared with wild-type controls, suggesting islet β-cell dysfunction. In summary, this study unravels a role for STK25 in determining the susceptibility to diet-induced non-alcoholic fatty pancreas disease in mice in connection to obesity. Our findings highlight STK25 as a potential drug target for metabolic disease.
Collapse
Affiliation(s)
- Esther Nuñez-Durán
- Department of Molecular and Clinical MedicineLundberg Laboratory for Diabetes Research, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Belén Chanclón
- Department of Molecular and Clinical MedicineLundberg Laboratory for Diabetes Research, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Metabolic PhysiologyInstitute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Silva Sütt
- Department of Molecular and Clinical MedicineLundberg Laboratory for Diabetes Research, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joana Real
- Department of Metabolic PhysiologyInstitute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical MedicineWallenberg Laboratory, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Metabolic PhysiologyInstitute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Emmelie Cansby
- Department of Molecular and Clinical MedicineLundberg Laboratory for Diabetes Research, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Margit Mahlapuu
- Department of Molecular and Clinical MedicineLundberg Laboratory for Diabetes Research, Institute of Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
26
|
Chursa U, Nuñez-Durán E, Cansby E, Amrutkar M, Sütt S, Ståhlman M, Olsson BM, Borén J, Johansson ME, Bäckhed F, Johansson BR, Sihlbom C, Mahlapuu M. Overexpression of protein kinase STK25 in mice exacerbates ectopic lipid accumulation, mitochondrial dysfunction and insulin resistance in skeletal muscle. Diabetologia 2017; 60:553-567. [PMID: 27981357 PMCID: PMC6518105 DOI: 10.1007/s00125-016-4171-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/14/2016] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Understanding the molecular networks controlling ectopic lipid deposition and insulin responsiveness in skeletal muscle is essential for developing new strategies to treat type 2 diabetes. We recently identified serine/threonine protein kinase 25 (STK25) as a critical regulator of liver steatosis, hepatic lipid metabolism and whole body glucose and insulin homeostasis. Here, we assessed the role of STK25 in control of ectopic fat storage and insulin responsiveness in skeletal muscle. METHODS Skeletal muscle morphology was studied by histological examination, exercise performance and insulin sensitivity were assessed by treadmill running and euglycaemic-hyperinsulinaemic clamp, respectively, and muscle lipid metabolism was analysed by ex vivo assays in Stk25 transgenic and wild-type mice fed a high-fat diet. Lipid accumulation and mitochondrial function were also studied in rodent myoblasts overexpressing STK25. Global quantitative phosphoproteomics was performed in skeletal muscle of Stk25 transgenic and wild-type mice fed a high-fat diet to identify potential downstream mediators of STK25 action. RESULTS We found that overexpression of STK25 in transgenic mice fed a high-fat diet increases intramyocellular lipid accumulation, impairs skeletal muscle mitochondrial function and sarcomeric ultrastructure, and induces perimysial and endomysial fibrosis, thereby reducing endurance exercise capacity and muscle insulin sensitivity. Furthermore, we observed enhanced lipid accumulation and impaired mitochondrial function in rodent myoblasts overexpressing STK25, demonstrating an autonomous action for STK25 within cells. Global phosphoproteomic analysis revealed alterations in the total abundance and phosphorylation status of different target proteins located predominantly to mitochondria and sarcomeric contractile elements in Stk25 transgenic vs wild-type muscle, respectively, providing a possible molecular mechanism for the observed phenotype. CONCLUSIONS/INTERPRETATION STK25 emerges as a new regulator of the complex interplay between lipid storage, mitochondrial energetics and insulin action in skeletal muscle, highlighting the potential of STK25 antagonists for type 2 diabetes treatment.
Collapse
Affiliation(s)
- Urszula Chursa
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Esther Nuñez-Durán
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Emmelie Cansby
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Manoj Amrutkar
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Silva Sütt
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Jan Borén
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Maria E Johansson
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bengt R Johansson
- Institute of Biomedicine, Electron Microscopy Unit, University of Gothenburg, Gothenburg, Sweden
| | - Carina Sihlbom
- Proteomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Margit Mahlapuu
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 5, SE-41345, Gothenburg, Sweden.
| |
Collapse
|
27
|
Amrutkar M, Kern M, Nuñez-Durán E, Ståhlman M, Cansby E, Chursa U, Stenfeldt E, Borén J, Blüher M, Mahlapuu M. Protein kinase STK25 controls lipid partitioning in hepatocytes and correlates with liver fat content in humans. Diabetologia 2016; 59:341-53. [PMID: 26553096 DOI: 10.1007/s00125-015-3801-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/13/2015] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is closely associated with pathological lipid accumulation in the liver, which is suggested to actively contribute to the development of insulin resistance. We recently identified serine/threonine protein kinase 25 (STK25) as a regulator of liver steatosis, whole-body glucose tolerance and insulin sensitivity in a mouse model system. The aim of this study was to assess the role of STK25 in the control of lipid metabolism in human liver. METHODS Intracellular fat deposition, lipid metabolism and insulin sensitivity were studied in immortalised human hepatocytes (IHHs) and HepG2 hepatocellular carcinoma cells in which STK25 was overexpressed or knocked down by small interfering RNA. The association between STK25 mRNA expression in human liver biopsies and hepatic fat content was analysed. RESULTS Overexpression of STK25 in IHH and HepG2 cells enhanced lipid deposition by suppressing β-oxidation and triacylglycerol (TAG) secretion, while increasing lipid synthesis. Conversely, knockdown of STK25 attenuated lipid accumulation by stimulating β-oxidation and TAG secretion, while inhibiting lipid synthesis. Furthermore, TAG hydrolase activity was repressed in hepatocytes overexpressing STK25 and reciprocally increased in cells with STK25 knockdown. Insulin sensitivity was reduced in STK25-overexpressing cells and enhanced in STK25-deficient hepatocytes. We also found a statistically significant positive correlation between STK25 mRNA expression in human liver biopsies and hepatic fat content. CONCLUSIONS/INTERPRETATION Our data suggest that STK25 regulates lipid partitioning in human liver cells by controlling TAG synthesis as well as lipolytic activity and thereby NEFA release from lipid droplets for β-oxidation and TAG secretion. Our findings highlight STK25 as a potential drug target for the prevention and treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Manoj Amrutkar
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Matthias Kern
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Esther Nuñez-Durán
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Emmelie Cansby
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Urszula Chursa
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden
| | - Elin Stenfeldt
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Margit Mahlapuu
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Blå stråket 5, SE-41345, Gothenburg, Sweden.
| |
Collapse
|
28
|
Amrutkar M, Cansby E, Chursa U, Nuñez-Durán E, Chanclón B, Ståhlman M, Fridén V, Mannerås-Holm L, Wickman A, Smith U, Bäckhed F, Borén J, Howell BW, Mahlapuu M. Genetic Disruption of Protein Kinase STK25 Ameliorates Metabolic Defects in a Diet-Induced Type 2 Diabetes Model. Diabetes 2015; 64:2791-804. [PMID: 25845663 PMCID: PMC4876789 DOI: 10.2337/db15-0060] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/31/2015] [Indexed: 12/17/2022]
Abstract
Understanding the molecular networks controlling ectopic lipid deposition, glucose tolerance, and insulin sensitivity is essential to identifying new pharmacological approaches to treat type 2 diabetes. We recently identified serine/threonine protein kinase 25 (STK25) as a negative regulator of glucose and insulin homeostasis based on observations in myoblasts with acute depletion of STK25 and in STK25-overexpressing transgenic mice. Here, we challenged Stk25 knockout mice and wild-type littermates with a high-fat diet and showed that STK25 deficiency suppressed development of hyperglycemia and hyperinsulinemia, improved systemic glucose tolerance, reduced hepatic gluconeogenesis, and increased insulin sensitivity. Stk25(-/-) mice were protected from diet-induced liver steatosis accompanied by decreased protein levels of acetyl-CoA carboxylase, a key regulator of both lipid oxidation and synthesis. Lipid accumulation in Stk25(-/-) skeletal muscle was reduced, and expression of enzymes controlling the muscle oxidative capacity (Cpt1, Acox1, Cs, Cycs, Ucp3) and glucose metabolism (Glut1, Glut4, Hk2) was increased. These data are consistent with our previous study of STK25 knockdown in myoblasts and reciprocal to the metabolic phenotype of Stk25 transgenic mice, reinforcing the validity of the results. The findings suggest that STK25 deficiency protects against the metabolic consequences of chronic exposure to dietary lipids and highlight the potential of STK25 antagonists for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Manoj Amrutkar
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Emmelie Cansby
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Urszula Chursa
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Esther Nuñez-Durán
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Belén Chanclón
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Vincent Fridén
- Center for Physiology and Bio-Imaging, Core Facilities, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Mannerås-Holm
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Anna Wickman
- Center for Physiology and Bio-Imaging, Core Facilities, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jan Borén
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Brian W Howell
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY
| | - Margit Mahlapuu
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
29
|
Amrutkar M, Cansby E, Nuñez-Durán E, Pirazzi C, Ståhlman M, Stenfeldt E, Smith U, Borén J, Mahlapuu M. Protein kinase STK25 regulates hepatic lipid partitioning and progression of liver steatosis and NASH. FASEB J 2015; 29:1564-76. [PMID: 25609431 DOI: 10.1096/fj.14-264937] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of liver disease, and 10% to 20% of NAFLD patients progress to nonalcoholic steatohepatitis (NASH). The molecular pathways controlling progression to NAFLD/NASH remain poorly understood. We recently identified serine/threonine protein kinase 25 (STK25) as a regulator of whole-body insulin and glucose homeostasis. This study investigates the role of STK25 in liver lipid accumulation and NASH. Stk25 transgenic mice challenged with a high-fat diet displayed a dramatic increase in liver steatosis and hepatic insulin resistance compared to wild-type siblings. Focal fibrosis, hepatocellular damage, and inflammation were readily seen in transgenic but not wild-type livers. Transgenic livers displayed reduced β-oxidation and triacylglycerol secretion, while lipid uptake and synthesis remained unchanged. STK25 was associated with lipid droplets, colocalizing with the main hepatic lipid droplet-coating protein adipose differentiation-related protein, the level of which was increased 3.8 ± 0.7-fold in transgenic livers (P < 0.01), while a key hepatic lipase, adipose triacylglycerol lipase, was translocated from the lipid droplets surface to the cytoplasm, providing the likely mechanism underlying the effect of STK25. In summary, STK25 is a lipid droplet-associated protein that promotes NAFLD through control of lipid release from the droplets for β-oxidation and triacylglycerol secretion. STK25 also drives pathogenesis of NASH.
Collapse
Affiliation(s)
- Manoj Amrutkar
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Emmelie Cansby
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Esther Nuñez-Durán
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Carlo Pirazzi
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Elin Stenfeldt
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Margit Mahlapuu
- *Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, and Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
30
|
Slubowski CJ, Paulissen SM, Huang LS. The GCKIII kinase Sps1 and the 14-3-3 isoforms, Bmh1 and Bmh2, cooperate to ensure proper sporulation in Saccharomyces cerevisiae. PLoS One 2014; 9:e113528. [PMID: 25409301 PMCID: PMC4237420 DOI: 10.1371/journal.pone.0113528] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/28/2014] [Indexed: 11/19/2022] Open
Abstract
Sporulation in the budding yeast Saccharomyces cerevisiae is a developmental program initiated in response to nutritional deprivation. Sps1, a serine/threonine kinase, is required for sporulation, but relatively little is known about the molecular mechanisms through which it regulates this process. Here we show that SPS1 encodes a bona-fide member of the GCKIII subfamily of STE20 kinases, both through phylogenetic analysis of the kinase domain and examination of its C-terminal regulatory domain. Within the regulatory domain, we find Sps1 contains an invariant ExxxPG region conserved from plant to human GCKIIIs that we call the EPG motif; we show this EPG motif is important for SPS1 function. We also find that Sps1 is phosphorylated near its N-terminus on Threonine 12, and that this phosphorylation is required for the efficient production of spores. In Sps1, Threonine 12 lies within a 14-3-3 consensus binding sequence, and we show that the S. cerevisiae 14-3-3 proteins Bmh1 and Bmh2 bind Sps1 in a Threonine 12-dependent fashion. This interaction is significant, as BMH1 and BMH2 are required during sporulation and genetically interact with SPS1 in sporulating cells. Finally, we observe that Sps1, Bmh1 and Bmh2 are present in both the nucleus and cytoplasm during sporulation. We identify a nuclear localization sequence in Sps1 at amino acids 411-415, and show that this sequence is necessary and sufficient for nuclear localization. Taken together, these data identify regions within Sps1 critical for its function and indicate that SPS1 and 14-3-3s act together to promote proper sporulation in S. cerevisiae.
Collapse
Affiliation(s)
- Christian J. Slubowski
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts, United States of America
| | - Scott M. Paulissen
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts, United States of America
| | - Linda S. Huang
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
Hwang J, Pallas DC. STRIPAK complexes: structure, biological function, and involvement in human diseases. Int J Biochem Cell Biol 2014; 47:118-48. [PMID: 24333164 PMCID: PMC3927685 DOI: 10.1016/j.biocel.2013.11.021] [Citation(s) in RCA: 185] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/18/2013] [Accepted: 11/28/2013] [Indexed: 12/31/2022]
Abstract
The mammalian striatin family consists of three proteins, striatin, S/G2 nuclear autoantigen, and zinedin. Striatin family members have no intrinsic catalytic activity, but rather function as scaffolding proteins. Remarkably, they organize multiple diverse, large signaling complexes that participate in a variety of cellular processes. Moreover, they appear to be regulatory/targeting subunits for the major eukaryotic serine/threonine protein phosphatase 2A. In addition, striatin family members associate with germinal center kinase III kinases as well as other novel components, earning these assemblies the name striatin-interacting phosphatase and kinase (STRIPAK) complexes. Recently, there has been a great increase in functional and mechanistic studies aimed at identifying and understanding the roles of STRIPAK and STRIPAK-like complexes in cellular processes of multiple organisms. These studies have identified novel STRIPAK and STRIPAK-like complexes and have explored their roles in specific signaling pathways. Together, the results of these studies have sparked increased interest in striatin family complexes because they have revealed roles in signaling, cell cycle control, apoptosis, vesicular trafficking, Golgi assembly, cell polarity, cell migration, neural and vascular development, and cardiac function. Moreover, STRIPAK complexes have been connected to clinical conditions, including cardiac disease, diabetes, autism, and cerebral cavernous malformation. In this review, we discuss the expression, localization, and protein domain structure of striatin family members. Then we consider the diverse complexes these proteins and their homologs form in various organisms, emphasizing what is known regarding function and regulation. Finally, we explore possible roles of striatin family complexes in disease, especially cerebral cavernous malformation.
Collapse
Affiliation(s)
- Juyeon Hwang
- Department of Biochemistry and Winship Cancer Institute, and Biochemistry, Cell, Developmental Biology Graduate Program, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA.
| | - David C Pallas
- Department of Biochemistry and Winship Cancer Institute, and Biochemistry, Cell, Developmental Biology Graduate Program, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA.
| |
Collapse
|
32
|
SOcK, MiSTs, MASK and STicKs: the GCKIII (germinal centre kinase III) kinases and their heterologous protein-protein interactions. Biochem J 2013; 454:13-30. [PMID: 23889253 DOI: 10.1042/bj20130219] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The GCKIII (germinal centre kinase III) subfamily of the mammalian Ste20 (sterile 20)-like group of serine/threonine protein kinases comprises SOK1 (Ste20-like/oxidant-stress-response kinase 1), MST3 (mammalian Ste20-like kinase 3) and MST4. Initially, GCKIIIs were considered in the contexts of the regulation of mitogen-activated protein kinase cascades and apoptosis. More recently, their participation in multiprotein heterocomplexes has become apparent. In the present review, we discuss the structure and phosphorylation of GCKIIIs and then focus on their interactions with other proteins. GCKIIIs possess a highly-conserved, structured catalytic domain at the N-terminus and a less-well conserved C-terminal regulatory domain. GCKIIIs are activated by tonic autophosphorylation of a T-loop threonine residue and their phosphorylation is regulated primarily through protein serine/threonine phosphatases [especially PP2A (protein phosphatase 2A)]. The GCKIII regulatory domains are highly disorganized, but can interact with more structured proteins, particularly the CCM3 (cerebral cavernous malformation 3)/PDCD10 (programmed cell death 10) protein. We explore the role(s) of GCKIIIs (and CCM3/PDCD10) in STRIPAK (striatin-interacting phosphatase and kinase) complexes and their association with the cis-Golgi protein GOLGA2 (golgin A2; GM130). Recently, an interaction of GCKIIIs with MO25 has been identified. This exhibits similarities to the STRADα (STE20-related kinase adaptor α)-MO25 interaction (as in the LKB1-STRADα-MO25 heterotrimer) and, at least for MST3, the interaction may be enhanced by cis-autophosphorylation of its regulatory domain. In these various heterocomplexes, GCKIIIs associate with the Golgi apparatus, the centrosome and the nucleus, as well as with focal adhesions and cell junctions, and are probably involved in cell migration, polarity and proliferation. Finally, we consider the association of GCKIIIs with a number of human diseases, particularly cerebral cavernous malformations.
Collapse
|
33
|
Cansby E, Amrutkar M, Mannerås Holm L, Nerstedt A, Reyahi A, Stenfeldt E, Borén J, Carlsson P, Smith U, Zierath JR, Mahlapuu M. Increased expression of STK25 leads to impaired glucose utilization and insulin sensitivity in mice challenged with a high-fat diet. FASEB J 2013; 27:3660-71. [PMID: 23729594 DOI: 10.1096/fj.13-228494] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Partial depletion of serine/threonine protein kinase 25 (STK25), a member of the Ste20 superfamily of kinases, increases lipid oxidation and glucose uptake in rodent myoblasts. Here we show that transgenic mice overexpressing STK25, when challenged with a high-fat diet, develop reduced glucose tolerance and insulin sensitivity compared to wild-type siblings, as evidenced by impairment in glucose and insulin tolerance tests as well as in euglycemic-hyperinsulinemic clamp studies. The fasting plasma insulin concentration was elevated in Stk25 transgenic mice compared to wild-type littermates (4.9±0.8 vs. 2.6±0.4 ng/ml after 17 wk on high-fat diet, P<0.05). Overexpression of STK25 decreased energy expenditure during the dark phase of observation (P<0.05), despite increased spontaneous activity. The oxidative capacity of skeletal muscle of transgenic carriers was reduced, as evidenced by altered expression of Cpt1, Acox1, and ACC. Hepatic triglycerides and glycogen were elevated (1.6- and 1.4-fold, respectively; P<0.05) and expression of key enzymes regulating lipogenesis (Fasn), glycogen synthesis (Gck), and gluconeogenesis (G6pc, Fbp1) was increased in the liver of the transgenic mice. Our findings suggest that overexpression of STK25 in conditions of excess dietary fuels associates with a shift in the metabolic balance in peripheral tissues from lipid oxidation to storage, leading to a systemic insulin resistance.
Collapse
Affiliation(s)
- Emmelie Cansby
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Blå stråket 5, SE-41345 Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|