5
Accepted Article This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1111/liv.12556 This article is protected by copyright. All rights reserved. Received Date : 11-Feb-2014 Accepted Date : 26-Mar-2014 Article type : Editorials MLK3 as a regulator of disease progression in NASH Joy X. Jiang and Natalie J. Török Department of Internal Medicine, Division of Gastroenterology and Hepatology UC Davis Medical Center, Sacramento, CA Address for correspondence: Natalie J. Török MD., UC Davis Medical Center, 4150 V Street, Suite 3500, Sacramento, CA 95817. Email: [email protected]; fax: 916-734-7908. Non-alcoholic steatohepatitis (NASH) is one of the most common chronic liver diseases worldwide (1). Despite the large number of studies published in the field, the molecular signals triggering the progression of NASH from simple steatosis to necroinflammation are still poorly understood. One of the most important and early features of progressive NASH is lipoapoptosis of hepatocytes that creates a proinflammatory and fibrogenic environment (2, 3). The activation of c-jun N-terminal kinase 1(JNK1) in both hepatocytes and macrophages has been described as a key event in NASH (4, 5), thus the signals governing its induction need to be better

MLK3 as a regulator of disease progression in Non-alcoholic steatohepatitis

Embed Size (px)

Citation preview

Acc

epte

d A

rtic

le

This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1111/liv.12556

This article is protected by copyright. All rights reserved.

Received Date : 11-Feb-2014

Accepted Date : 26-Mar-2014

Article type : Editorials

MLK3 as a regulator of disease progression in NASH

Joy X. Jiang and Natalie J. Török

Department of Internal Medicine, Division of Gastroenterology and Hepatology UC Davis

Medical Center, Sacramento, CA

Address for correspondence:

Natalie J. Török MD., UC Davis Medical Center, 4150 V Street, Suite 3500, Sacramento, CA

95817. Email: [email protected]; fax: 916-734-7908.

Non-alcoholic steatohepatitis (NASH) is one of the most common chronic liver diseases

worldwide (1). Despite the large number of studies published in the field, the molecular signals

triggering the progression of NASH from simple steatosis to necroinflammation are still poorly

understood. One of the most important and early features of progressive NASH is lipoapoptosis

of hepatocytes that creates a proinflammatory and fibrogenic environment (2, 3). The activation

of c-jun N-terminal kinase 1(JNK1) in both hepatocytes and macrophages has been described

as a key event in NASH (4, 5), thus the signals governing its induction need to be better

Acc

epte

d A

rtic

le

This article is protected by copyright. All rights reserved.

elucidated. The paper published by Ibrahim et al (6) has focused on the role of the mixed-

lineage kinase 3 (MLK3) as an important proximal JNK activator that has a major role in

progressive liver injury in diet-induced NASH. Several studies demonstrated that MLK3/JNK

activation plays an essential role in saturated fatty acid-induced insulin resistance and

hepatocyte lipotoxicity (7-9). Exposure to free fatty acid induced MLK3/JNK in mouse

embryonic fibroblasts, and the MLK3-/- cells displayed increased insulin sensitivity (8); the same

trend was found in the MLK3-/- mice on high fat diet (8, 10). In hepatocytes, palmitate induced

the recruitment of cdc42/Rac1 and the activation of MLK3/JNK, leading to downstream ER

stress signaling (7). In a different study, the palmitate-induced M1 macrophage polarization was

diminished in the MLK3-/- cells (10). All of these suggest that MLK3/JNK signaling plays an

important role in the progression of NASH.

MLKs are MAPK-kinase kinases (MKKKs) thatactivate JNK and p38 signaling cascades via the

MAPK kinases (MKKs) by either forming a homodimer or associating with Rho GTPases (11-

13).The selective activation of MKKs and the downstream induction of JNK1/2 or p38 by MLKs

are mediated by the MAPK scaffold proteins such as JNK interacting proteins (JIPs) (13, 14).

How these JIPs direct selective activation of their targets is still under investigation. JIP3 was

shown to bind to MLK3/MKK7 inducing JNK1 activation in neurons (14-16). It is however, not

clear which JIPs are predominant in the liver and whether they selectively mediate JNK1 and 2

activation.

JNK activationplays a key role in saturated fatty acid-induced hepatocyte apoptosis(5), both in

humans and in animal models (17-19). In HSC, phosphorylated-JNK1 directly mediates

transdifferentiation contributing to fibrosis (20). MLK3/JNK/P38 activation in LX-2 cells has

been observed when cells were stimulated with a PPARβ/δ ligand, leading to cell proliferation

(21). JNK1 and JNK2 have differential effects in NASH: JNK1 mediates steatohepatitis and

lipotoxicity, whereas JNK2 activation is more protective (22, 23). It is not yet known whether

Acc

epte

d A

rtic

le

This article is protected by copyright. All rights reserved.

MLK3 has differential effects on JNK1 or JNK2 in the liver, and if so, how the differential

induction is mediated. The current study is in agreement with previous findings demonstrating

that the MLK3-/- mice developed less hepatic steatosis, decreased liver injury, inflammation and

fibrosis. The diet used in this study mimics the fast food diet consumed by humans inducing

insulin resistance, steatohepatitis and fibrosis (24). It has been recently reported that compared

to the WT mice, the global MLK3-deficient mice on high fat diet displayed significantly less

weight gain, and reduced macrophage infiltration in the adipose tissue and also decreased

systemic inflammation (10). Interestingly, these mice had increased energy expenditure that

could have accounted for the slower weight gain. The improved inflammation in the liver could

be explained by the decreased macrophage recruitment and JNK inactivation in the MLK3-/-

mice (10). Using the MLK2/3 double knockout mice on the high fat diet model, Davis and

colleagues showed that the obesity-resistant phenotype of these animals was related to the

upregulation of the sympatho-adrenal system, as using a selective antagonist to the β3

adrenergic receptor prevented the increase in body temperature and decreased the expression

of the adrenergic target genes (25). Distinct from the studies described above, the authors in

the current study chose high fat diet combined with high fructose consumption. The mice in this

study gained similar amount of weight in both treatment arms, and this could be attributed to the

high fructose intake. As fibrosis was diminished in the MLK3-/- mice it is likely that MLK3 in

stellate cells was involved in their transdifferentiation, which has been observed in lung

fibroblasts by Lin et al (26). Macrophage polarization could also be affected by MLK3 (10); and

there is evidence showing that MLK3 interacts with TLR signaling by directly binding to Myd88

(27). The dominant cell type and mechanisms for the proinflammatory and fibrogenic activity of

MLK3 in the liver would require further investigation, and could be addressed in the future by the

generation of conditional knockout mice.

Acc

epte

d A

rtic

le

This article is protected by copyright. All rights reserved.

References:

1. Lazo M, Clark JM. The epidemiology of nonalcoholic fatty liver disease: a global perspective. Semin Liver Dis 2008;28:339-350. 2. Malhi H, Bronk SF, Werneburg NW, Gores GJ. Free fatty acids induce JNK-dependent hepatocyte lipoapoptosis. J Biol Chem 2006;281:12093-12101. 3. Farrell GC, van Rooyen D, Gan L, Chitturi S. NASH is an Inflammatory Disorder: Pathogenic, Prognostic and Therapeutic Implications. Gut Liver 2012;6:149-171. 4. Han MS, Jung DY, Morel C, Lakhani SA, Kim JK, Flavell RA, Davis RJ. JNK expression by macrophages promotes obesity-induced insulin resistance and inflammation. Science 2013;339:218-222. 5. Ibrahim SH, Gores GJ. Who pulls the trigger: JNK activation in liver lipotoxicity? J Hepatol 2012;56:17-19. 6. Ibrahim SH, Gores GJ, Hirsova P, Kirby M, Miles L, Jaeschke A, Kohli R. Mixed lineage kinase 3 deficient mice are protected against the high fat high carbohydrate diet-induced steatohepatitis. Liver Int 2013. 7. Sharma M, Urano F, Jaeschke A. Cdc42 and Rac1 are major contributors to the saturated fatty acid-stimulated JNK pathway in hepatocytes. J Hepatol 2012;56:192-198. 8. Jaeschke A, Davis RJ. Metabolic stress signaling mediated by mixed-lineage kinases. Mol Cell 2007;27:498-508. 9. Leamy AK, Egnatchik RA, Young JD. Molecular mechanisms and the role of saturated fatty acids in the progression of non-alcoholic fatty liver disease. Prog Lipid Res 2013;52:165-174. 10. Gadang V, Kohli R, Myronovych A, Hui DY, Perez-Tilve D, Jaeschke A. MLK3 promotes metabolic dysfunction induced by saturated fatty acid-enriched diet. Am J Physiol Endocrinol Metab 2013;305:E549-556. 11. Leung IW, Lassam N. Dimerization via tandem leucine zippers is essential for the activation of the mitogen-activated protein kinase kinase kinase, MLK-3. J Biol Chem 1998;273:32408-32415. 12. Teramoto H, Coso OA, Miyata H, Igishi T, Miki T, Gutkind JS. Signaling from the small GTP-binding proteins Rac1 and Cdc42 to the c-Jun N-terminal kinase/stress-activated protein kinase pathway. A role for mixed lineage kinase 3/protein-tyrosine kinase 1, a novel member of the mixed lineage kinase family. J Biol Chem 1996;271:27225-27228. 13. Gallo KA, Johnson GL. Mixed-lineage kinase control of JNK and p38 MAPK pathways. Nat Rev Mol Cell Biol 2002;3:663-672. 14. Yasuda J, Whitmarsh AJ, Cavanagh J, Sharma M, Davis RJ. The JIP group of mitogen-activated protein kinase scaffold proteins. Mol Cell Biol 1999;19:7245-7254. 15. Kelkar N, Gupta S, Dickens M, Davis RJ. Interaction of a mitogen-activated protein kinase signaling module with the neuronal protein JIP3. Mol Cell Biol 2000;20:1030-1043. 16. Kelkar N, Standen CL, Davis RJ. Role of the JIP4 scaffold protein in the regulation of mitogen-activated protein kinase signaling pathways. Mol Cell Biol 2005;25:2733-2743. 17. Puri P, Mirshahi F, Cheung O, Natarajan R, Maher JW, Kellum JM, Sanyal AJ. Activation and dysregulation of the unfolded protein response in nonalcoholic fatty liver disease. Gastroenterology 2008;134:568-576. 18. Schattenberg JM, Nagel M, Kim YO, Kohl T, Worns MA, Zimmermann T, Schad A, et al. Increased hepatic fibrosis and JNK2-dependent liver injury in mice exhibiting hepatocyte-specific deletion of cFLIP. Am J Physiol Gastrointest Liver Physiol 2012;303:G498-506.

Acc

epte

d A

rtic

le

This article is protected by copyright. All rights reserved.

19. Wang Y, Ausman LM, Russell RM, Greenberg AS, Wang XD. Increased apoptosis in high-fat diet-induced nonalcoholic steatohepatitis in rats is associated with c-Jun NH2-terminal kinase activation and elevated proapoptotic Bax. J Nutr 2008;138:1866-1871. 20. Zhao G, Hatting M, Nevzorova YA, Peng J, Hu W, Boekschoten MV, Roskams T, et al. Jnk1 in murine hepatic stellate cells is a crucial mediator of liver fibrogenesis. Gut 2013. 21. Kostadinova R, Montagner A, Gouranton E, Fleury S, Guillou H, Dombrowicz D, Desreumaux P, et al. GW501516-activated PPARbeta/delta promotes liver fibrosis via p38-JNK MAPK-induced hepatic stellate cell proliferation. Cell Biosci 2012;2:34. 22. Singh R, Wang Y, Xiang Y, Tanaka KE, Gaarde WA, Czaja MJ. Differential effects of JNK1 and JNK2 inhibition on murine steatohepatitis and insulin resistance. Hepatology 2009;49:87-96. 23. Schattenberg JM, Singh R, Wang Y, Lefkowitch JH, Rigoli RM, Scherer PE, Czaja MJ. JNK1 but not JNK2 promotes the development of steatohepatitis in mice. Hepatology 2006;43:163-172. 24. Charlton M, Krishnan A, Viker K, Sanderson S, Cazanave S, McConico A, Masuoko H, et al. Fast food diet mouse: novel small animal model of NASH with ballooning, progressive fibrosis, and high physiological fidelity to the human condition. Am J Physiol Gastrointest Liver Physiol 2011;301:G825-834. 25. Kant S, Barrett T, Vertii A, Noh YH, Jung DY, Kim JK, Davis RJ. Role of the mixed-lineage protein kinase pathway in the metabolic stress response to obesity. Cell Rep 2013;4:681-688. 26. Lin CH, Yu MC, Tung WH, Chen TT, Yu CC, Weng CM, Tsai YJ, et al. Connective tissue growth factor induces collagen I expression in human lung fibroblasts through the Rac1/MLK3/JNK/AP-1 pathway. Biochim Biophys Acta 2013;1833:2823-2833. 27. Sun D, Ding A. MyD88-mediated stabilization of interferon-gamma-induced cytokine and chemokine mRNA. Nat Immunol 2006;7:375-381.