Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 

REVIEW ARTICLE

The Role of Stearoyl-coenzyme A Desaturase 1 in Liver 
Development, Function, and Pathogenesis
Fatemeh Mohammadzadeh1, Vahid Hosseini2, Alireza Alihemmati3, Maghsod Shaaker1, 
Gholamali Mosayyebi4, Masoud Darabi1, Amir Mehdizadeh5

1Emergency Medicine Research Center Team, Department of  Emergency Medicine, Tabriz University of  Medical Sciences, Tabriz, Iran; 
2 Department of  Biochemistry and Clinical Laboratories, Faculty of  Medicine, Tabriz University of  Medical Sciences, Tabriz, Iran; 
3 Department of  Anatomical Sciences, Faculty of  Medicine, Tabriz University of  Medical Sciences, Tabriz, Iran; 4Liver and Gastrointestinal 
Research Center, Tabriz University of  Medical Sciences, Tabriz, Iran; 5Endocrine Research Center, Tabriz University of  Medical Sciences, 
Tabriz, Iran

Abstract

Stearoyl-coenzyme A desaturase 1 (SCD1) is a microsomal enzyme that controls fatty acid metabolism and is highly expressed in 
hepatocytes. SCD1 may play a key role in liver development and hepatic lipid homeostasis through promoting monounsaturated 
protein acylation and converting lipotoxic saturated fatty acids into monounsaturated fatty acids. Imbalanced activity of  SCD1 
has been implicated in fatty liver induction, inflammation and stress. In this review, the role of  SCD1 in hepatic development, 
function and pathogenesis is discussed. Additionally, emerging novel therapeutic agents targeting SCD1 for the treatment of  liver 
disorders are presented.

Keywords: hepatic lipogenesis; hydroxy pyridine; MK-8245; stearoyl-coenzyme A desaturase 1; SCD1

Received: 20 December 2018; Accepted after Revision: 15 January 2019; Published: 06 February 2019

Authors for correspondence: Masoud Darabi, Emergency Medicine Research Center Team, Department of  Emergency Medicine, 
Tabriz University of  Medical Sciences, Tabriz, Iran. Email: darabim@tbzmed.ac.ir; Amir Mehdizadeh, Endocrine Research Center, Tabriz 
University of  Medical Sciences, Tabriz, Iran. Email: mehdizadeha@tbzmed.ac.ir

How to cite: Mohammadzadeh F et al. The role of  Stearoyl-coenzyme A  Desaturase 1 in liver development, function and pathogenesis. J Ren 
Hepat Disord. 2019;3(1):15–22.

Doi: http://dx.doi.org/10.15586/jrenhep.2019.49

Copyright: Mohammadzadeh F et al.

License: This open access article is licensed under Creative Commons Attribution 4.0 International (CC BY 4.0).  
http://creativecommons.org/licenses/by/4.0

Introduction
Stearoyl-coenzyme A desaturase 1 (SCD1) was discovered in 
1988 when Ntambi and colleagues identified an mRNA tran-
script whose expression was highly induced during adipogenic 
differentiation (1). SCD1 is an iron-containing lipid- regulating 
enzyme that is highly expressed in the liver and is the main en-
zyme responsible for de novo synthesis of  monounsaturated 

fatty acids (MUFAs). Palmitoyl-CoA and stearoyl-CoA are 
the main substrates of  this enzyme, converting them into 
palmitoleoyl-CoA and oleoyl-CoA, respectively (2).

SCD1 is coded by its gene on the long arm of  chromosome 
24, in the sub-band 3 of  region 24 (3). Promoter  activity  region 
is located within the initial 609 bp upstream of  transcription 
initiation site which constitutes a CCAAT-box identified as 

P   U   B   L   I   C   A   T   I   O   N   S
 CODON

Journal of Renal and Hepatic Disorders

mailto:darabim@tbzmed.ac.ir
mailto:mehdizadeha@tbzmed.ac.ir
http://dx.doi.org/10.15586/jrenhep.2019.49
http://creativecommons.org/licenses/by/4.0


Mohammadzadeh F et al.

 Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 16

a cis-element binding site. Sterol regulatory element-binding 
transcription factor 1 (SREBP-1c), liver X receptor (LXR), 
peroxisome proliferator-activated receptor alpha (PPAR-α) 
and CCAAT/enhancer-binding protein alpha (C/EBP-α) are 
among the most important transcription factors that bind 
to SCD1 promoter and control its gene expression (4). The 
pseudogene of  SCD1, containing two premature stop codons 
downstream of  the original start codon, is located on the short 
arm of  chromosome 24 in the sub-band 32 of  region 11 (5).

SCD1 protein is a microsomal enzyme containing four 
transmembrane domains in which both the N-terminus and 
C-terminus are located in the cytoplasm (Figure 1). Eight his-
tidine residues on the single cytoplasmic loop and C- terminus 
are conserved and important for desaturase catalytic activ-
ity (6). Purified SCD1 protein migrates as a 37 kDa band by 
SDS gel electrophoresis (7, 8). As fatty acids are important 
components of  phospholipids, triglycerides and esterified 
cholesterol, changes in SCD1 expression and activity can af-
fect membrane stability, lipid metabolism and the amount of  
adipose tissue; consequent changes may be associated with 
obesity, fatty liver, cancers, diabetes and atherosclerosis (9). 
This review provides an overview of  the role of  SCD1 on var-
ious aspects of  liver pathophysiology such as development, 
hepatic lipogenesis and inflammation. It also summarizes 
the role of  novel small molecules targeting SCD1 as potential 
agents for the treatment of  various liver disorders.

SCD1 Activity Contributes to Liver Development 
through Protein Acylation
The Wnt family of  proteins are signaling molecules that 
orchestrate numerous homeostatic events from embryonic 
development to adult tissue function (10). Their malfunc-
tion causes various hepatic abnormalities. The products of  
SCD1 can regulate Wnt trafficking and function through 
MUFA acylation or lipidation (Figure 2). Monounsaturated 
fatty acyl moieties render Wnts hydrophobic and insoluble 
in aqueous environment (11). During the embryonic stage, 
the inner layer, endoderm, is partitioned into three regions 
termed as foregut, midgut, and hindgut, with the foregut 
containing liver precursors. The liver and biliary tracts de-
velop from the foregut at the 4th week of  gestation (12). The 
intermediate germ layer, mesoderm, produces Wnt which 
contributes to the development of  hindgut in the posterior 
endoderm. In the anterior endoderm, however, suppression 
of  Wnt signaling retains foregut fate and allows subsequent 
development of  the liver (13, 14). Overall, Wnt signaling is 
tightly regulated during embryo development, which is par-
ticularly important at the initial stages of  liver development. 
Absence of  Wnt signaling activity will result in impaired he-
patic development. This is supported by the elevated expres-
sion of  Wnt downstream core transcription factors during 
the terminal differentiation of  hepatocytes (15).

SCD1 shows a determinant role in the in vitro differentia-
tion process of  human-induced pluripotent stem cells toward 
hepatic lineage. Inhibition of  SCD1 by a selective inhibitor 
in early stages of  in vitro induced differentiation has resulted 

Figure 1.  Stearoyl-coenzyme A desaturase 1 (SCD1) 
is an iron-containing transmembrane enzyme. SCD1 
protein is  exclusively localized on the ER membrane 
with both the N- and C-terminal domains stretched into 
the cytosol. It has four transmembrane helices (purple 
cylindrical shapes). The eight histidines (hexagonal shapes) 
on the single cytoplasmic loop and C-terminus are highly 
conserved, particularly regions surrounding the  di-iron  
center. The cytosolic domain provides a structural frame for 
the regioselectivity and stereospecificity of the desaturation 
reaction (6).

Figure 2.  Stearoyl-coenzyme A desaturase 1 (SCD1) 
contributes to liver development and regeneration by 
modulating Wnt activity. Microsomal SCD1 produces 
monounsaturated fatty acids that can be attached to 
Wnts. This process, termed acylation, enables Wnt secretion 
and activation. Wnt acylation is also a prerequisite for the 
formation of  concentration and activity gradients of  Wnts. 
The Wnt gradients mediate zonal development of  liver and 
regeneration. 



Stearoyl-coenzyme A Desaturase 1 and liver 

 Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 17

in decreased hepatic markers. In the rescue experiments with 
the combination of  the SCD1 inhibitor and its main product 
oleate, the effect of  inhibitor was strongly reversed (16). Our 
ongoing in vivo work is to identify the role of  SCD1 in the 
early stages of  liver organogenesis and development, espe-
cially its role in the formation of  a complete liver before birth.

Apart from hepatocyte differentiation, Wnt signaling path-
way plays a central role in liver zonation (17). Hepatocytes 
 adjacent to portal vein are named periportal hepatocytes 
(zone 1) and involved in glyconeogenesis and β-oxidation, 
whereas those near the central vein are called pericentral hepa-
tocytes (zone 3) and play a distinct role in drug metabolism 
and glycolysis. Hepatocytes in zone 2 exhibit an intermediary 
role having both periportal and pericentral functions. In a re-
cent study, a high β-catenin-dependent Wnt signaling activity 
was observed in zone 3 (17–19). These findings collectively 
highlight the importance of  the SCD1- produced MUFAs in 
 mediating liver development through Wnt regulation.

Recent studies using animal models of  partial hepatec-
tomy have also implicated the direct involvement of  Wnt 
signaling pathway in liver regeneration. The expression and 
nuclear translocation of  β-catenin significantly increase 
within minutes of  hepatectomy and promote hepatocyte pro-
liferation rate (20). In acetaminophen-induced liver injury, 
Wnt/β- catenin not only induces the expression of  enzymes 
involved in drug metabolism such as cytochrome P450 2E1 
and cytochrome P450 1A2 but also helps liver regeneration 
(21, 22). In line with this finding, SCD1 gene expression was 
increased 3.5-fold in  regenerating liver following major liver 
resection (23). After tissue damage, the residual cells start 
the repair process (24), and the proliferating cells accumulate 
several categories of  lipids  including triglycerides, fatty acids 
(both saturated and unsaturated), and cholesterol esters (25). 
These lipids are thought to undergo oxidation and provide 
the required energy for cell proliferation. In addition, they 
are the main components of  newly synthetized membranes. 
Besides, these lipids can contribute to various signal trans-
duction cascades related to cell proliferation and differentia-
tion such as Wnt and hedgehog signaling pathways (26). It is 
hypothesized that MUFAs produced by SCD1 not only pro-
vide metabolic energy source and structural components, but 
also promote protein acylation–mediated signaling, which 
are essential for liver regeneration.

SCD1 Prevents Lipotoxicity and Controls Hepatic 
Lipogenesis
Palmitic acid and stearic acid are the major de novo synthe-
sized lipotoxic saturated fatty acids (SFAs) in the liver. SCD1 
mediates the addition of  a double bond to the saturated 
carbon chain. On one hand, SCD1 activity attenuates SFAs 
lipotoxic effects through their conversion into the MUFAs 
palmitoleic acid and oleic acid (27). On the other hand, SCD1 
generates unsaturated fatty acids serving as rate-limiting 

substrates for lipogenesis (28). Thus, an improper increase in 
its activity may cause hepatic lipid accumulation.

Despite oleate being the major dietary MUFA, SCD1 
expression is highly regulated in response to developmen-
tal, dietary, environmental, and hormonal factors. De novo 
synthesized MUFAs are the preferred substrates for neutral 
hepatic lipid synthesis including triglycerides and cholesterol 
ester. SCD1 inhibition protects against high-fat high-carbo-
hydrate diet, leptin-deficiency-induced obesity, and hepatic 
steatosis (29). Leptin-deficient mice exhibit SCD1 overex-
pression causing palmitoleate and oleate accumulation in 
liver as fat droplets. Recent studies on high-carbohydrate-fed 
rats have revealed that deficiency in SCD1 decreased lipid 
synthesis, elevated fatty acid oxidation and thermogenesis, 
and insulin susceptibility in different tissues, especially in the 
liver (30).

Sampath et al. (31) showed that stearate-rich diet causes 
SCD1 induction and hepatic lipid accumulation in wild-type 
mice but not in scd1-/- mice. However, in scd1-/- mice, stearate 
does not induce genes involved in lipogenesis. Additionally, 
sterol regulatory element-binding protein-1c (SREBP-1c) 
and peroxisome proliferator-activated receptor coactiva-
tor-1 (PGC-1) transcription factors, which are necessary 
mediators for pro-lipogenic activities of  saturated fatty acids 
(SFAs), were downregulated in scd1-/- mice. Instead, fatty 
acid oxidation genes such as carnitine palmitoyltransferase-1 
(CPT-1) were induced, resulting in hepatic glycogen deple-
tion. Lui et  al. (32), in a study on zinc finger transcription 
factor knockout mouse model, showed a significant decrease 
in SCD1 expression and triglyceride accumulation compared 
to controls. Binding of  this transcription factor to SCD1 pro-
moter in hepatocytes was also reported in this study, indicat-
ing its critical role in the activation of  SCD1 expression.

The role of  SCD1 in gut microbiota-dependent hepatic 
lipogenesis has been studied by Singh et al. (28). They re-
ported that mice with deficient Toll-like receptor-5, which is 
expressed in gut epithelial cells and plays an important role 
in microbiota homeostasis, exhibit a microbiota-dependent 
metabolic syndrome with elevated hepatic lipogenesis, SCD1 
expression and activity, and hepatic neutral lipids accumula-
tion with a high oleate and palmitoleate content.

Furthermore, the expression level of  SCD1 and fatty 
acid synthase along with endoplasmic reticulum (ER) stress 
markers was downregulated in response to the inhibition of  
poly ADP-ribose polymerase (PARP), which is overexpressed 
in long-term high-fat high-sucrose diet in mice, indicating the 
PARP-SCD1 interaction as a major mechanism in the in-
duction of  non-alcoholic fatty liver disease (33). The role of  
SCD1 in an alcoholic fatty liver disease model was studied by 
Louinis et al. (34). In that study, mice fed with a low-MUFA 
diet containing 5% ethanol for 10 days and a single ethanol 
gavage (5 g/kg) developed severe hepatic injury. Liver-specific 
Scd1-knock-out (SCD1-LKO) mice were resistant to such 
hepatic injury.



Mohammadzadeh F et al.

 Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 18

In a recent cross-sectional clinical study, it has been shown 
that the serum SCD1 activity index is significantly related to 
the risk of  non-alcoholic fatty liver disease in patients with 
primary dyslipidemia (35). Of  course, while interpreting 
these data, it should be noted that the index of  serum SCD1 
activity is not solely determined by the liver. In this field, 
the Ntambi laboratory has recently shown that hepatic tri-
glyceride accumulation in mice may be induced by increased 
hepatic trafficking of  MUFAs originating from non-liver 
 tissues (36). The latter finding further supports the hypoth-
esis that fine-tuning of  hepatic SCD1 activity is critical in 
variable metabolic states.

SCD1 Modulates Hepatic Inflammation and 
Oxidative Stress
Current evidence indicates the regulatory role of  fatty acids 
in cellular inflammation. SCD1 plays an important role in 
maintaining the balance between SFAs and MUFAs. Toxic 
accumulation of  SFAs that are reflected as MUFAs/SFAs 
imbalance leads to activation of  oxidative stress imbalance 
in hepatocytes (37, 38). SFAs mediate cellular inflammatory 
response through binding to Toll-like receptor-4, CD14, and 
myeloid differentiation protein-2, causing increased produc-
tion of  bacterial lipopolysaccharides, oxidized phospholip-
ids, and oxidized low-density lipoproteins through intestinal 
microbiota modification (39). These findings support the hy-
pothesis that SCD1 activity may be protective against SFA-
induced oxidative stress and hepatic inflammation. Lu et al. 
(40) reported that IKK2 (an activator of  NFκB) activation 
can induce hepatic SCD1 overexpression and triglyceride 
accumulation in mouse. However, such activation decreased 
the expression of  oxidative stress and prevented hepatic in-
flammation and fibrosis. Paradoxically, there is evidence that 
increased SCD1 activity may contribute to inflammation and 
oxidative stress. In the high-carbohydrate or high-sucrose, 
very-low-fat diet, oleate supplementation leads to decreased 
hepatic injury and oxidative stress in mice with liver-specific 
SCD1-LKO (41). A high-fructose diet in female C57BL/6J 
mice also induced oxidative stress characterized by hepatic 
SCD1 overexpression and elevation of  inducible nitric oxide 
synthase levels (42). Ochi et al. (43) also showed the associa-
tion of  SCD1 with the development of  non-alcoholic steato-
hepatitis (NASH) under induced stress condition. Indeed, a 
knockout C57BL/6 mice model with a low expression and 
activity of  SCD1 showed lower hepatic lipid accumulation 
and steatosis following tunicamycin-induced ER stress than 
the wild-type mice with ER stress.

Overall, as illustrated in Figure 3, balanced activity of  
SCD1 is important for stabilizing the ratio of  unsaturated to 
saturated fatty acids. An increase in this ratio can lead to lipid 
accumulation and a decrease in this ratio is associated with 
lipotoxicity. A rise in both lipid accumulation and lipotox-
icity may lead to hepatic inflammation and oxidative stress.

SCD-1 as a Potential Therapeutic Target
The aforementioned studies highlight the significant role 
of  SCD1 activity in hepatic pathophysiology. Therapeutic 
strategies targeting SCD1 may have applications in man-
aging liver disorders. Several studies have examined small 
molecule inhibitors in this regard. The following sections 
review recent advances in small molecule SCD1 inhibitors 
and their potential therapeutic application in hepatic disor-
ders (Table 1).

MK-8245

SCD1 enzyme is expressed in many cell types of  the body. 
Therefore, potential SCD1 inhibitors for the treatment of  
liver diseases will have a lot of  side effects although they 
might be highly selective toward SCD1. For example, SCD1- 
knockout rodent models and SCD1 inhibitor–treated rats de-
velop severe skin and eye abnormalities (44, 45). Therefore, 
liver-specific targeting of  SCD1 may be an effective strategy 
for the treatment of  liver-related disorders. One such in-
hibitor is MK-8245 (46). It has a transporting element that 
specifically interacts with heptocytes via the liver-specific 
organic anion transporting polypeptides. Administration 
of  MK-8245 to mice fed with a high-fat diet did not reduce 
food intake. Despite this, a reduction in liver steatosis and a 
decrease in liver triglyceride levels were observed. MK-8245 
also exhibited anti-diabetic and anti-dyslipidemic proper-
ties. Administration of  MK-8245 to individuals with type 
2  diabetes mellitus in a phase II clinical trial showed no se-
rious adverse events (47). MK-8245 may also be an option 
for anti-hepatitis C virus (HCV) therapy as evaluated using 
recombinant HCV culture systems (48). The potential thera-
peutic effects of  this compound on liver diseases are yet to be 
clinically examined.

Figure 3.  Stearoyl-coenzyme A desaturase 1 (SCD1) 
activity is associated with normal liver function. The 
schematic balance represents that the equilibrium between 
saturated fatty acids (SFAs) and monounsaturated fatty acids 
(MUFAs) is important in the physiological state. The loss 
of  this equilibrium is due to overactivity and underactivity 
of  SCD1 in hepatic lipid accumulation and  lipotoxicity, 
respectively.



Stearoyl-coenzyme A Desaturase 1 and liver 

 Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 19

Hydroxy pyridone

Another compound that was used for targeted liver SCD1 
inhibition is 4-hydroxy pyridine (49). According to the 
pharmacokinetic analysis, this SCD1 inhibitor had a sig-
nificantly higher concentration in liver than in plasma and 
eyelid. It showed a good potency in reducing the mouse liver 
ratio of  palmitoleate to palmitate as a biomarker for SCD1 
activity (49).

Pyridazine derivative

A piperazine-based SCD-1 inhibitor, N-(2-hydroxy-2-
phenylethyl)-6-[4-(2 methylbenzoyl)piperidin-1-yl]pyridaz-
ine-3-carboxamide, has been shown to produce beneficial 
effects in experimental modes of  NASH (50, 51). When 
administered orally for 8 weeks, once daily, triglyceride 
accumulation in the liver was reduced by 80% from the 
fourth week. It also attenuated the increase of  aspartate 
aminotransferase and alanine transaminase by 86% and 
78%, respectively. Hepatic steatosis, hepatocellular degen-
eration, and inflammatory cell infiltration were also amelio-
rated after the treatment (50).

Thiazole analogs

Current evidence shows that host cell lipid homeostasis 
plays a critical role in the pathogenesis of  HCV by facilitat-
ing the formation of  viral membrane-associated replication 
complex. Since inhibiting lipogenesis has a negative effect 
on virus proliferation, inhibiting the lipid synthesis enzyme 
SCD1 is a potential strategy for HCV treatment (52, 53). Lyn 
et al. (54) showed that the thiazole compound MF-152 can 
repress HCV infection in human hepatoma cells by modify-
ing membrane functions which are required for HCV rep-
lication. Cell imaging studies showed that the inability of  
viral RNAs to interact with modified membranes exposes 
them to degradation by endogenous nucleases. This process 

ultimately prevents the formation of  HCV viral complexes 
and arrests HCV replication.

Thiazole-4-acetic acid derivative

Another potent liver-selective SCD1 inhibitor, compound 
48 (a thiazole-4-acetic acid derivative), has recently been 
discovered through high-throughput screening efforts fol-
lowing an ex vivo assay approach on mice liver and eyelids 
(55). Administration of  compound 48 to mice fed a high-
fat diet for 43 days improved glucose tolerance and de-
creased body weight without adverse effects on skin or eyes. 
Furthermore, compound 48 significantly attenuated hepatic 
triglyceride accumulation in rats fed a high-sucrose, very-
low-fat diet (55). These findings suggest that compound 48 
may have clinical benefits in the treatment of  diabetes, he-
patic steatosis, and obesity through targeting SCD1 activity 
in liver.

Conclusion
SCD1, one of  the predominantly expressed enzymes in the 
liver, is a major factor in fatty acid metabolism. It plays 
a regulatory role in posttranslational protein modifica-
tions via protein acylation. Wnts play an important role 
in  hepatic differentiation, zonation, and regeneration. 
The Wnts pathway, at least in part, is regulated by SCD1-
mediated palmitoleoylation and oleoylation. In addition, 
SCD1 is crucial for SFA detoxification and MUFA produc-
tion. SFAs overproduction induces hepatic inflammation 
and oxidative stress, and SCD1 attenuates these effects via 
monounsaturation of  SFAs. Meanwhile, de novo produced 
MUFAs promote and participate in cellular lipogenesis 
and their overproduction can lead to hepatic lipid accu-
mulation. Targeting SCD1 as a novel therapeutic approach 
may be beneficial in liver disorders. In this regard, several 
studies have tested small molecule inhibitors of  SCD1 

Table 1. Stearoyl-coenzyme A desaturase 1 small molecule inhibitors with potential applications for hepatic diseases.

Compound Liver-selective Therapeutic application Current evidence Reference

MK-8245 + Diabetes, dyslipidemia, 
HCV

Preclinical/animal/
phase II clinical trial

(46, 48)

Hydroxy pyridone + Dyslipidemia- In vivo (49)

N-(2-hydroxy-2-phenylethyl)-6-[4-
(2-methylbenzoyl)piperidin-1-yl]
pyridazine-3-carboxamide

- Reduction in triglyceride 
accumulation in NASH

In vivo (50, 51)

Thiazole analogs - HCV infection In vitro (54)

Thiazol-4-acetic acid derivatives + Diabetes, hepatic steatosis 
and obesity

In vivo (55)

HCV, hepatitis C virus; NASH, nonalcoholic steatohepatitis; OATP, organic-anion-transporting polypeptide.



Mohammadzadeh F et al.

 Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 20

in vitro and in vivo, which opens a promising point of  view 
in the treatment of  liver metabolic diseases.

Acknowledgments
This study was supported by a grant (No. 5.4.9705) from the 
Emergency Medicine Research Team at Tabriz University of  
Medical Sciences.

Conflict of interest
The authors declare no potential conflicts of  interest with 
respect to research, authorship, and/or publication of  this 
article.

References
 1.  Ntambi JM, Buhrow SA, Kaestner KH, Christy RJ, Sibley E, 

Kelly TJ, et al. Differentiation-induced gene expression in 3T3-
L1 preadipocytes. Characterization of  a differentially expressed 
gene encoding stearoyl-CoA desaturase. J Biol Chem [Internet]. 
1988 Nov 25;263(33):17291–300. Available from (accessed 
25.12.18.): http://www.ncbi.nlm.nih.gov/pubmed/2903162

 2.  Flowers MT, Ntambi JM. Role of  stearoyl-coenzyme A 
 desaturase in regulating lipid metabolism. Curr Opin Lipidol. 
2008 Jun;19(3):248–56. https://doi.org/10.1097/MOL.0b013e 
3282f9b54d

 3.  Arregui M, Buijsse B, Stefan N, Corella D, Fisher E, di 
Giuseppe R, et al. Heterogeneity of  the Stearoyl-CoA de-
saturase-1 (SCD1) gene and metabolic risk factors in the 
EPIC-Potsdam study. PLoS One. 2012;7:e48338. https://doi.
org/10.1371/ journal.pone.0048338

 4.  Mauvoisin D, Charfi  C, Lounis AM, Rassart E, Mounier 
C. Decreasing stearoyl-CoA desaturase-1 expression inhib-
its  β- catenin signaling in breast cancer cells. Cancer Sci. 2013 
Jan;104(1):36–42. Available from: https://doi.org/10.1111/
cas.12032

 5.  Zhang L, Ge L, Tran T, Stenn K, Prouty SM. Isolation and 
characterization of  the human stearoyl-CoA desaturase gene 
promoter: Requirement of  a conserved CCAAT cis-element. 
Biochem J. 2001;357:183–93. https://doi.org/10.1042/bj3570183

 6.  Bai Y, McCoy JG, Levin EJ, Sobrado P, Rajashankar KR, Fox 
BG, et al. X-ray structure of  a mammalian stearoyl-CoA de-
saturase. Nature. 2015 Aug;524(7564): 252–6. Available from: 
https://doi.org/10.1038/nature14549

 7.  Mehdizadeh A, Fayezi S, Darabi M. SCD (stearoyl-
CoA desaturase (delta-9-desaturase)). Atlas Genet 
Cytogenet Oncol Haematol. 2016;20(2):77–80. https://doi.
org/10.4267/2042/62515

 8.  Kim YC, Gomez FE, Fox BG, Ntambi JM. Differential reg-
ulation of  the stearoyl-CoA desaturase genes by thiazolidin-
ediones in 3T3-L1 adipocytes. J Lipid Res [Internet]. 2000 
Aug;41(8):1310–16. Available from (accessed 25.12.18.): http://
www.ncbi.nlm.nih.gov/pubmed/10946019

 9.  Miyazaki M, Dobrzyn A, Man WC, Chu K, Sampath H, 
Kim H-J, et al. Stearoyl-CoA Desaturase 1 gene expression 
is necessary for fructose-mediated induction of  lipogenic 
gene expression by sterol regulatory element-binding protein-
1c- dependent and -independent mechanisms. J Biol Chem. 
2004 Jun;279(24):25164–71. https://doi.org/10.1074/jbc. M40 
2781200

 10.  Steinhart Z, Angers S. Wnt signaling in development and tis-
sue homeostasis. Development. 2018;145(11):1–8. https://doi.
org/10.1242/dev.146589

 11.  Janda CY, Waghray D, Levin AM, Thomas C, Garcia KC. 
Structural basis of  Wnt recognition by Frizzled. Science. 2012 
Jul;337(6090):59–64. https://doi.org/10.1126/science.1222879

 12.  Sekine K, Takebe T, Taniguchi H. Liver regeneration using cul-
tured liver bud. Methods Mol Biol. 2017;1597:207–16. https://
doi.org/10.1007/978-1-4939-6949-4_15

 13.  Gualdi R, Bossard P, Zheng M, Hamada Y, Coleman JR, 
Zaret KS. Hepatic specification of  the gut endoderm in vitro: 
Cell signaling and transcriptional control. Genes Dev. 1996 
Jul;10(13):1670–82. https://doi.org/10.1101/gad.10.13.1670

 14.  Lokmane L, Haumaitre C, Garcia-Villalba P, Anselme I, 
Schneider-Maunoury S, Cereghini S. Crucial role of  vHNF1 
in vertebrate hepatic specification. Development. 2008 
Aug;135(16):2777–86. https://doi.org/10.1242/dev.023010

 15.  Lemaigre FP. Mechanisms of  liver development: Concepts 
for understanding liver disorders and design of  novel ther-
apies. Gastroenterology. 2009 Jul;137(1):62–79. https://doi.
org/10.1053/j.gastro.2009.03.035

 16.  Rahimi Y, Mehdizadeh A, Nozad Charoudeh H, Nouri M, 
Valaei K, Fayezi S, et al. Hepatocyte differentiation of  human 
induced pluripotent stem cells is modulated by stearoyl-CoA de-
saturase 1 activity. Dev Growth Differ. 2015 Dec;57(9):667–74. 
Available from: https://doi.org/10.1111/dgd.12255

 17.  Schröder E, Höhme S, Böttger J, Aleithe S, Gebhardt R,  Matz-Soja 
M. Zonation of  Morphogens in the adult liver – Crosstalk be-
tween Hh and Wnt/β-Catenin signaling. Z Gastroenterol. 2018 
Jan;56(01):E2–89. https://doi.org/10.1055/s-0037-1612748

 18. Brosch M, Kattler K, Herrmann A, von Schönfels W, 
Nordström  K, Seehofer D, et al. Epigenomic map of  human 
liver reveals principles of  zonated morphogenic and meta-
bolic control. Nat Commun. 2018 Oct;9(1):4150. https://doi.
org/10.1038/s41467-018-06611-5

 19. Preziosi M, Okabe H, Poddar M, Singh S, Monga SP. 
Endothelial Wnts regulate β-catenin signaling in murine liver 
zonation and regeneration: A sequel to the Wnt-Wnt situation. 
Hepatol Commun. 2018 Jul;2(7):845–60. https://doi.org/10.1002/
hep4.1196

 20. Nelsen CJ, Rickheim DG, Timchenko NA, Stanley MW, 
Albrecht JH. Transient expression of  cyclin D1 is sufficient to 
promote hepatocyte replication and liver growth in vivo. Cancer 
Res. 2001 Dec;61(23):8564–8.

 21. Yu X, Wang Y, DeGraff  DJ, Wills ML, Matusik RJ. Wnt/β-
catenin activation promotes prostate tumor progression in a 
mouse model. Oncogene. 2011 Apr;30(16):1868–79. https://doi.
org/10.1038/onc.2010.560

 22. Dadhania VP, Bhushan B, Apte U, Mehendale HM. Wnt/β-
Catenin signaling drives thioacetamide-mediated heteroprotec-
tion against acetaminophen-induced lethal liver injury. Dose 
Response. 15(1):1559325817690287. https://doi.org/10.1177/ 
1559325817690287

 23.  Xu C, Lin F, Qin S. Relevance between lipid metabolism- 
associated  genes and rat liver regeneration. Hepatol Res. 
2008  Aug;38(8): 825–37. https://doi.org/10.1111/j.1872-034X. 
2008. 00345.x

 24.  Forbes SJ, Newsome PN. Liver regeneration—mechanisms and 
models to clinical application. Nat Rev Gastroenterol Hepatol. 
2016;13(8):473–85. https://doi.org/10.1038/nrgastro.2016.97

 25.  Bellet MM, Masri S, Astarita G, Sassone-Corsi P, Della Fazia 
MA, Servillo G. Histone deacetylase SIRT1 controls prolif-
eration, circadian rhythm, and lipid metabolism during liver 

http://www.ncbi.nlm.nih.gov/pubmed/2903162
https://doi.org/10.1097/MOL.0b013e3282f9b54d
https://doi.org/10.1097/MOL.0b013e3282f9b54d
https://doi.org/10.1371/journal.pone.0048338
https://doi.org/10.1371/journal.pone.0048338
https://doi.org/10.1111/cas.12032
https://doi.org/10.1111/cas.12032
https://doi.org/10.1042/bj3570183
https://doi.org/10.1038/nature14549
https://doi.org/10.4267/2042/62515
https://doi.org/10.4267/2042/62515
http://www.ncbi.nlm.nih.gov/pubmed/10946019
http://www.ncbi.nlm.nih.gov/pubmed/10946019
https://doi.org/10.1074/jbc.M402781200
https://doi.org/10.1074/jbc.M402781200
https://doi.org/10.1242/dev.146589
https://doi.org/10.1242/dev.146589
https://doi.org/10.1126/science.1222879
https://doi.org/10.1007/978-1-4939-6949-4_15
https://doi.org/10.1007/978-1-4939-6949-4_15
https://doi.org/10.1101/gad.10.13.1670
https://doi.org/10.1242/dev.023010
https://doi.org/10.1053/j.gastro.2009.03.035
https://doi.org/10.1053/j.gastro.2009.03.035
https://doi.org/10.1111/dgd.12255
https://doi.org/10.1055/s-0037-1612748
https://doi.org/10.1038/s41467-018-06611-5
https://doi.org/10.1038/s41467-018-06611-5
https://doi.org/10.1002/hep4.1196
https://doi.org/10.1002/hep4.1196
https://doi.org/10.1038/onc.2010.560
https://doi.org/10.1038/onc.2010.560
https://doi.org/10.1177/1559325817690287
https://doi.org/10.1177/1559325817690287
https://doi.org/10.1111/j.1872-034X.2008.00345.x
https://doi.org/10.1111/j.1872-034X.2008.00345.x
https://doi.org/10.1038/nrgastro.2016.97


Stearoyl-coenzyme A Desaturase 1 and liver 

 Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 21

regeneration in mice. J Biol Chem. 2016;291(44):23318–29. 
https://doi.org/10.1074/jbc.M116.737114

 26.  Kohjima M, Tsai T-H, Tackett BC, Thevananther S, Li L, 
Chang BH-J, et al. Delayed liver regeneration after partial hepa-
tectomy in adipose differentiation related protein-null mice. J 
Hepatol. 2013 Dec;59(6):1246–54. https://doi.org/10.1016/j.
jhep.2013.07.025

 27.  Collins JM, Neville MJ, Hoppa MB, Frayn KN. De novo 
lipogenesis and stearoyl-CoA desaturase are coordinately reg-
ulated in the human adipocyte and protect against palmitate- 
induced cell injury. J Biol Chem. 2010;285:6044–52. https://doi.
org/10.1074/jbc.M109.053280

 28.  Singh V, Chassaing B, Zhang L, San Yeoh B, Xiao X, 
Kumar  M, et al. Microbiota-dependent hepatic lipogenesis 
mediated by Stearoyl CoA Desaturase 1 (SCD1) promotes 
metabolic syndrome in TLR5-deficient mice. Cell Metab. 2015 
Dec;22(6):983–96. https://doi.org/10.1016/j.cmet.2015.09.028

 29.  Miyazaki M, Sampath H, Liu X, Flowers MT, Chu K, 
Dobrzyn  A, et al. Stearoyl-CoA desaturase-1 deficiency atten-
uates obesity and insulin resistance in leptin-resistant obese 
mice. Biochem Biophys Res Commun. 2009 Mar;380(4):818–22. 
https://doi.org/10.1016/j.bbrc.2009.01.183

 30.  Miyazaki M, Flowers MT, Sampath H, Chu K, Otzelberger C, 
Liu X, et al. Hepatic stearoyl-CoA desaturase-1 deficiency 
protects mice from carbohydrate-induced adiposity and he-
patic steatosis. Cell Metab. 2007 Dec;6(6):484–96. https://doi.
org/10.1016/j.cmet.2007.10.014

 31.  Sampath H, Miyazaki M, Dobrzyn A, Ntambi JM. Stearoyl-
CoA desaturase-1 mediates the pro-lipogenic effects of  di-
etary  saturated fat. J Biol Chem. 2007 Jan;282(4): 2483–93. 
Available from: https://doi.org/10.1074/jbc.M610158200

 32.  Liu G, Zhou L, Zhang H, Chen R, Zhang Y, Li L, et al. 
Regulation of  hepatic lipogenesis by the zinc finger pro-
tein Zbtb20. Nat Commun. 2017 Mar;8:14824. https://doi.
org/10.1038/ncomms14824

 33.  Gariani K, Ryu D, Menzies KJ, Yi H-S, Stein S, Zhang  H, 
et  al. Inhibiting poly ADP-ribosylation increases fatty 
acid oxidation and protects against fatty liver disease. 
J Hepatol. 2017;66(1):132–41. https://doi.org/10.1016/j.jhep. 
2016.08.024

 34.  Lounis MA, Escoula Q, Veillette C, Bergeron K-F, Ntambi JM, 
Mounier C. SCD1 deficiency protects mice against ethanol- 
induced liver injury. Biochim Biophys Acta. 2016;1861(11): 
1662–70. https://doi.org/10.1016/j.bbalip.2016.07.012

 35.  Amor AJ, Cofán M, Mateo-Gallego R, Cenarro A, Civeira F, 
Ortega E, et al. Dietary polyunsaturated fatty acids mediate the 
inverse association of  stearoyl-CoA desaturase activity with the 
risk of  fatty liver in dyslipidaemic individuals. Eur J Nutr. 2018 
Apr; Available from: https://doi.org/10.1007/s00394-018-1691-4

 36.  Bond LM, Ntambi JM. UCP1 deficiency increases adipose 
tissue monounsaturated fatty acid synthesis and trafficking to 
the liver. J Lipid Res. 2018 Feb;59(2):224–36. Available from: 
https://doi.org/10.1194/jlr.M078469

 37.  Wek RC, Anthony TG. Obesity: Stressing about unfolded pro-
teins. Nat Med. 2010 Apr;16(4):374–6. https://doi.org/10.1038/
nm0410-374

 38.  Ron D, Walter P. Signal integration in the endoplasmic retic-
ulum unfolded protein response. Nat Rev Mol Cell Biol. 2007 
Jul;8(7):519–29. https://doi.org/10.1038/nrm2199

 39.  Rocha DM, Caldas AP, Oliveira LL, Bressan J, Hermsdorff  
HH. Saturated fatty acids trigger TLR4-mediated inflamma-
tory response. Atherosclerosis. 2016 Jan;244:211–5. https://doi.
org/10.1016/j.atherosclerosis.2015.11.015

 40.  Lu H, Lei X, Zhang Q. Moderate activation of  IKK2-NF-kB in 
unstressed adult mouse liver induces cytoprotective genes and 
lipogenesis without apparent signs of  inflammation or fibrosis. 
BMC Gastroenterol. 2015 Jul;15:94. https://doi.org/10.1186/
s12876-015-0325-z

 41.  Liu X, Burhans MS, Flowers MT, Ntambi JM. Hepatic oleate 
regulates liver stress response partially through PGC-1α dur-
ing high-carbohydrate feeding. J Hepatol. 2016;65(1):103–12. 
https://doi.org/10.1016/j.jhep.2016.03.001

 42.  Choi Y, Abdelmegeed MA, Song B-J. Diet high in fructose pro-
motes liver steatosis and hepatocyte apoptosis in C57BL/6J fe-
male mice: Role of  disturbed lipid homeostasis and increased 
oxidative stress. Food Chem Toxicol. 2017 May;103:111–21. 
https://doi.org/10.1016/j.fct.2017.02.039

 43.  Ochi T, Munekage K, Ono M, Higuchi T, Tsuda M, Hayashi Y, 
et al. Patatin-like phospholipase domain-containing protein 3 
is involved in hepatic fatty acid and triglyceride metabolism 
through X-box binding protein 1 and modulation of  endo-
plasmic reticulum stress in mice. Hepatol Res. 2016 May;46(6): 
584–92. https://doi.org/10.1111/hepr.12587

 44.  Miyazaki M, Man WC, Ntambi JM. Targeted disruption of  
stearoyl-CoA desaturase1 gene in mice causes atrophy of  seba-
ceous and meibomian glands and depletion of  wax esters in the 
eyelid. J Nutr. 2001 Sep;131(9):2260–8. Available from: https://
doi.org/10.1093/jn/131.9.2260

 45.  Ramtohul YK, Black C, Chan C-C, Crane S, Guay J, Guiral S, 
et  al. SAR and optimization of  thiazole analogs as potent 
stearoyl-CoA desaturase inhibitors. Bioorg Med Chem Lett. 
2010 Mar;20(5):1593–7. https://doi.org/10.1016/j.bmcl.2010. 
01.083

 46.  Oballa RM, Belair L, Black WC, Bleasby K, Chan CC, 
Desroches C, et al. Development of  a liver-targeted stearoyl-
CoA desaturase (SCD) inhibitor (MK-8245) to establish a ther-
apeutic window for the treatment of  diabetes and dyslipidemia. 
J Med Chem. 2011 Jul;54(14):5082–96. https://doi.org/10.1021/
jm200319u

 47.  Merck Sharp & Dohme Corp. A study to assess the safety 
and efficacy of  MK8245 in patients with type 2 diabetes mel-
litus and inadequate glycemic control (MK8245-005 AM2) 
[Internet]. ClinicalTrials.gov Web Site. 2019. Available from 
(accessed 25.12.18.): https://clinicaltrials.gov/ct2/show/study/
NCT00846391

 48.  Nio Y, Hasegawa H, Okamura H, Miyayama Y, Akahori Y, 
Hijikata M. Liver-specific mono-unsaturated fatty acid syn-
thase-1 inhibitor for anti-hepatitis C treatment. Antiviral Res. 
2016 Aug;132:262–7. Available from: https://doi.org/10.1016/j.
antiviral.2016.07.003

 49.  Sun S, Zhang Z, Raina V, Pokrovskaia N, Hou D,  
Namdari R, et al. Discovery of  thiazolylpyridinone SCD1  
inhibitors with preferential liver distribution and reduced 
mechanism-based adverse effects. Bioorg Med Chem Lett. 
2014 Jan;24(2):526–31. https://doi.org/10.1016/j.bmcl. 2013. 
12.035

 50.  Uto Y, Ogata T, Kiyotsuka Y, Ueno Y, Miyazawa Y, Kurata H, 
et al. Novel benzoylpiperidine-based stearoyl-CoA desatu-
rase-1 inhibitors: Identification of  6-[4-(2-methylbenzoyl)
piperidin-1-yl]pyridazine-3-carboxylic acid (2-hydroxy-2-pyrid-
in-3-ylethyl)amide and its plasma triglyceride-lowering effects in 
Zucker fatty rats. Bioorg Med Chem Lett. 2010 Jan;20(1):341–5. 
https://doi.org/10.1016/j.bmcl.2009.10.101

 51.  Kurikawa N, Takagi T, Wakimoto S, Uto Y, Terashima H, 
Kono K, et al. A novel inhibitor of  stearoyl-CoA desaturase-1 at-
tenuates hepatic lipid accumulation, liver injury and inflammation 

https://doi.org/10.1074/jbc.M116.737114
https://doi.org/10.1016/j.jhep.2013.07.025
https://doi.org/10.1016/j.jhep.2013.07.025
https://doi.org/10.1074/jbc.M109.053280
https://doi.org/10.1074/jbc.M109.053280
https://doi.org/10.1016/j.cmet.2015.09.028
https://doi.org/10.1016/j.bbrc.2009.01.183
https://doi.org/10.1016/j.cmet.2007.10.014
https://doi.org/10.1016/j.cmet.2007.10.014
https://doi.org/10.1074/jbc.M610158200
https://doi.org/10.1038/ncomms14824
https://doi.org/10.1038/ncomms14824
https://doi.org/10.1016/j.jhep.2016.08.024
https://doi.org/10.1016/j.jhep.2016.08.024
https://doi.org/10.1016/j.bbalip.2016.07.012
https://doi.org/10.1007/s00394-018-1691-4
https://doi.org/10.1194/jlr.M078469
https://doi.org/10.1038/nm0410-374
https://doi.org/10.1038/nm0410-374
https://doi.org/10.1038/nrm2199
https://doi.org/10.1016/j.atherosclerosis.2015.11.015
https://doi.org/10.1016/j.atherosclerosis.2015.11.015
https://doi.org/10.1186/s12876-015-0325-z
https://doi.org/10.1186/s12876-015-0325-z
https://doi.org/10.1016/j.jhep.2016.03.001
https://doi.org/10.1016/j.fct.2017.02.039
https://doi.org/10.1111/hepr.12587
https://doi.org/10.1093/jn/131.9.2260
https://doi.org/10.1093/jn/131.9.2260
https://doi.org/10.1016/j.bmcl.2010.01.083
https://doi.org/10.1016/j.bmcl.2010.01.083
https://doi.org/10.1021/jm200319u
https://doi.org/10.1021/jm200319u
https://clinicaltrials.gov/ct2/show/study/NCT00846391
https://clinicaltrials.gov/ct2/show/study/NCT00846391
https://doi.org/10.1016/j.antiviral.2016.07.003
https://doi.org/10.1016/j.antiviral.2016.07.003
https://doi.org/10.1016/j.bmcl.2013.12.035
https://doi.org/10.1016/j.bmcl.2013.12.035
https://doi.org/10.1016/j.bmcl.2009.10.101


Mohammadzadeh F et al.

 Journal of Renal and Hepatic Disorders 2019; 3(1): 15–22 22

in model of  nonalcoholic steatohepatitis. Biol Pharm Bull. 
2013;36(2):259–67. https://doi.org/10.1248/bpb.b12-00702

 52.  Pezacki JP, Singaravelu R, Lyn RK. Host-virus interactions 
during hepatitis C virus infection: A complex and dynamic mo-
lecular biosystem. Mol Biosyst. 2010 Jul;6(7):1131–42. https://
doi.org/10.1039/b924668c

 53.  Herker E, Ott M. Unique ties between hepatitis C virus repli-
cation and intracellular lipids. Trends Endocrinol Metab. 2011 
Jun;22(6):241–8. Available from: https://doi.org/10.1016/j.tem. 
2011. 03.004

 54.  Lyn RK, Singaravelu R, Kargman S, O’Hara S, Chan H, Oballa 
R, et al. Stearoyl-CoA desaturase inhibition blocks formation 
of  hepatitis C virus-induced specialized membranes. Sci Rep. 
2014 Apr;4:4549. https://doi.org/10.1038/srep04549

 55.  Iida T, Ubukata M, Mitani I, Nakagawa Y, Maeda K, Imai H, 
et  al. Discovery of  potent liver-selective stearoyl-CoA desatu-
rase-1 (SCD1) inhibitors, thiazole-4-acetic acid derivatives, for 
the treatment of  diabetes, hepatic steatosis, and obesity. Eur 
J Med Chem. 2018 Oct;158:832–52. https://doi.org/10.1016/j.
ejmech. 2018.09.003

https://doi.org/10.1248/bpb.b12-00702
https://doi.org/10.1039/b924668c
https://doi.org/10.1039/b924668c
https://doi.org/10.1016/j.tem.2011.03.004
https://doi.org/10.1016/j.tem.2011.03.004
https://doi.org/10.1038/srep04549
https://doi.org/10.1016/j.ejmech.2018.09.003
https://doi.org/10.1016/j.ejmech.2018.09.003