ArticlePDF AvailableLiterature Review

Mitochondrial CPT1A: Insights into structure, function, and basis for drug development

Frontiers
Frontiers in Pharmacology
Authors:

Abstract and Figures

Carnitine Palmitoyl-Transferase1A (CPT1A) is the rate-limiting enzyme in the fatty acid β-oxidation, and its deficiency or abnormal regulation can result in diseases like metabolic disorders and various cancers. Therefore, CPT1A is a desirable drug target for clinical therapy. The deep comprehension of human CPT1A is crucial for developing the therapeutic inhibitors like Etomoxir. CPT1A is an appealing druggable target for cancer therapies since it is essential for the survival, proliferation, and drug resistance of cancer cells. It will help to lower the risk of cancer recurrence and metastasis, reduce mortality, and offer prospective therapy options for clinical treatment if the effects of CPT1A on the lipid metabolism of cancer cells are inhibited. Targeted inhibition of CPT1A can be developed as an effective treatment strategy for cancers from a metabolic perspective. However, the pathogenic mechanism and recent progress of CPT1A in diseases have not been systematically summarized. Here we discuss the functions of CPT1A in health and diseases, and prospective therapies targeting CPT1A. This review summarizes the current knowledge of CPT1A, hoping to prompt further understanding of it, and provide foundation for CPT1A-targeting drug development.
Mitochondrial CPT1A: Insights
into structure, function, and basis
for drug development
Kai Liang*
School of Life Science, Peking University, Beijing, China
Carnitine Palmitoyl-Transferase1A (CPT1A) is the rate-limiting enzyme in the fatty
acid β-oxidation, and its deciency or abnormal regulation can result in diseases
like metabolic disorders and various cancers. Therefore, CPT1A is a desirable drug
target for clinical therapy. The deep comprehension of human CPT1A is crucial for
developing the therapeutic inhibitors like Etomoxir. CPT1A is an appealing
druggable target for cancer therapies since it is essential for the survival,
proliferation, and drug resistance of cancer cells. It will help to lower the risk
of cancer recurrence and metastasis, reduce mortality, and offer prospective
therapy options for clinical treatment if the effects of CPT1A on the lipid
metabolism of cancer cells are inhibited. Targeted inhibition of CPT1A can be
developed as an effective treatment strategy for cancers from a metabolic
perspective. However, the pathogenic mechanism and recent progress of
CPT1A in diseases have not been systematically summarized. Here we discuss
the functions of CPT1A in health and diseases, and prospective therapies targeting
CPT1A. This review summarizes the current knowledge of CPT1A, hoping to
prompt further understanding of it, and provide foundation for CPT1A-
targeting drug development.
KEYWORDS
CPT1a, FATTY ACID β-OXIDATION, cancer, inhibitor, drug development
Introduction
Globally, the prevalence of obesity has increased, and more and more attention has been
paid to lipid metabolism (Bluher, 2019). Fatty acid oxidation (FAO), a process that occurs
within cells, is an important energy source. Diabetes patients cannot use sugar as their
primary source of energy since they have poor insulin sensitivity (Shepherd and Kahn, 1999).
Additionally, the majority of cancer cells use lipid metabolism as a source of energy (Munir
et al., 2019). Reducing or inhibiting FAO can turn off the energy source of cancer cells and
starve them to death (Bergers and Fendt, 2021).
The majority of FAO happens in mitochondrion. Long chain fatty acids (LCFAs) cannot
directly enter the mitochondrial inner membrane, and CPT1A is required to convert acyl-
CoA (carbon chain longer than 12) into acyl-carnitine (Ceccarelli et al., 2011). CACT
(carnitine/acylcarnitine carrier protein), a transporter in the inner mitochondrial
membrane, transports free carnitine out of the mitochondrial matrix and into the
cytoplasm as well as acyl-carnitine into the mitochondrial matrix (Indiveri et al., 2011).
Acyl-carnitine entering the mitochondrial matrix is again converted to acyl-CoA by
CPT2 and participates in the true fatty acid β-oxidation cycle (Indiveri et al., 2011).
OPEN ACCESS
EDITED BY
Jing Wu,
Shandong Provincial Qianfoshan
Hospital, China
REVIEWED BY
Chunming Cheng,
The Ohio State University, United States
Chao Sun,
The Second Hospital of Shandong
University, China
*CORRESPONDENCE
Kai Liang,
liangkai@pku.edu.cn
SPECIALTY SECTION
This article was submitted to
Experimental Pharmacology and Drug
Discovery,
a section of the journal
Frontiers in Pharmacology
RECEIVED 07 February 2023
ACCEPTED 13 March 2023
PUBLISHED 23 March 2023
CITATION
Liang K (2023), Mitochondrial CPT1A:
Insights into structure, function, and basis
for drug development.
Front. Pharmacol. 14:1160440.
doi: 10.3389/fphar.2023.1160440
COPYRIGHT
© 2023 Liang. This is an open-access
article distributed under the terms of the
Creative Commons Attribution License
(CC BY). The use, distribution or
reproduction in other forums is
permitted, provided the original author(s)
and the copyright owner(s) are credited
and that the original publication in this
journal is cited, in accordance with
accepted academic practice. No use,
distribution or reproduction is permitted
which does not comply with these terms.
Frontiers in Pharmacology frontiersin.org01
TYPE Review
PUBLISHED 23 March 2023
DOI 10.3389/fphar.2023.1160440
CPT1A is a rate-limiting enzyme of FAO that catalyzes the
transfer of the long-chain acyl group in acyl-CoA ester to carnitine,
allowing fatty acids to enter the mitochondrial matrix for oxidation
(Figure 1)(Houten et al., 2016).
CPT1 was originally identied by McGarry and Foster in
1978, and they hypothesized that CPT1 is the rate-limiting
enzyme in fatty acid oxidation (McGarry et al., 1978). CPT1A
can be divided into three isoforms, known as CPT1A, CPT1B,
and CPT1C, based on its tissue distribution, and sequence
characteristics. CPT1A is extensively expressed in the liver,
kidney, pancreas, adipose tissue, lymphocytes, and broblast,
while CPT1B and CPT1C have strict tissue-specic
distributions (Bonnefontetal.,2004). CPT1A has a higher
afnity for carnitine (Km = 30 μM for ratCPT1A) than CPT1B
(Km = 500 μMforratCPT1B)(Ramsay et al., 2001;Ceccarelli
et al., 2011). Both CPT1A and CPT1B have signicant effects on
the metabolic syndrome, cardiovascular disease, type
2 diabetes, and other disorders (Bonnefontetal.,2004).
Human CPT1A and CPT1B share 63% of their total
sequence homology, 82% around the active region (Figure 2)
(Ceccarelli et al., 2011). CPT1C is mostly found in the
hypothalamus and hippocampus, where it can control
ceramide levels and inuence learning, cognition (Virmani
et al., 2015). Although CPT1C can bind malonyl-CoA, it has
low catalytic activity, making functional investigations difcult
FIGURE 1
Chart showing how the CPT shuttle system transports palmitoyl-
CoA into the mitochondria. The carnitine shuttle system includes
CPT1A, CACT, and CPT2. Mitochondrial fatty acid β-oxidation (FAO) is
started by the successive actions of CPT1A (in the outer
membrane) and CPT2 (in the inner membrane), together with a
carnitine-acylcarnitine translocase (CACT).
FIGURE 2
Multiple sequence alignment was performed by ClustalW in MEGA11 software (Tamura et al., 2021) and further treated by ESPript webserver (Robert
and Gouet, 2014). Sequence alignment of three CPT1 isoforms with strictly conserved amino acid residues highlighted in box.
Frontiers in Pharmacology frontiersin.org02
Liang 10.3389/fphar.2023.1160440
(Fado et al., 2021). Spastic paraplegia, which is exclusive to
CPT1C and has nothing in common with CPT1A deciency, is
most likely caused by a dominant genetic variant in CPT1C
(Rinaldi et al., 2015).
CPT1A gene encodes a protein with 773 amino acids, which is a
transmembrane protein located in the outer membrane of
mitochondria facing the cytoplasm (Ramsay et al., 2001). Two
transmembrane helical regions divide CPT1A protein into the
small N-terminal regulatory region (about 47 amnio acids) and
the main C-terminal catalytic domain (Gobin et al., 2003). Near the
N-terminal region of CPT1A, two transmembrane helices act as an
anchor to the mitochondrial outer membrane. The majority of the
N- and C-termini are located on the cytoplasmic side, leaving just a
loop of about 27 amino acid residues at the N-terminus in the inner
and outer membrane space (Figure 3)(Fraser et al., 1997).
The basic state of CPT1A is a trimer, which has the potential to
further merge into a hexamer, and may play a role in the sensitivity
to malonyl-CoA (Faye et al., 2007). CPT1A, together with acyl-CoA
synthetase (ACSL) and voltage-dependent anion channel (VDAC)
form the fatty acid transfer complex located in the outer membrane
(Lee et al., 2011).
So far, it is difcult to nd a membrane (or membrane-like)
condition that can maintain the enzyme activity of CPT1 to make
the protein soluble, let alone to study the structure of this enzyme.
Despite characterization of the ratCPT2 crystal structure, the
sequence similarity with CPT1A is low, making it of little use as
a reference for CPT1A structure research (Gobin et al., 2003).
Structure of CPT1A
CPT1A was discovered in 1978 as the result of a gene located at
11q13.3 that comprises 22 exons and belongs to the carnitine/
choline acetyltransferase family (McGarry et al., 1978). The
CPT1A gene codes for a 773 amino acid protein that has a
short N-terminal regulatory domain (residues 147), a
mitochondrial intermembrane domain (residues 74102), two
transmembrane (TM) domains (residues 4873 for TM1 and
residues 103122 for TM2), and a catalytic domain (residues
123773). A conformation shift in the N-terminal region is
critical in malonyl-CoA-mediated enzyme activity control (Rao
et al., 2011).
CPT1A can be homo-oligomerized to form trimers (Figure 4),
which further form hexamers (Faye et al., 2007;Jenei et al., 2009).
It has been suggested that the interaction between the GXXXG
and GXXXA motifs in CPT1A TM2 helix is essential for
its oligomerization (Jenei et al., 2009). Another study revealed
that long chain acyl-CoA synthetase 1 (ACSL1) and the voltage-
dependent anion channel (VDAC) were also immunocaptured
by CPT1A antibodies, suggesting that CPT1A, ACSL1, and
VDAC may all be members of mitochondrial outer
membrane acylcarnitine translocation complex (Lee et al.,
2011). Regarding the oligomerization of CPT1A, both models
have a justication for existing due to the lack of structural
support, and further (in situ) structural studies are needed to
resolve this issue.
FIGURE 3
Molecular properties and physiological functions of CPT1A. (A) Domain structure of the full-length CPT1A; Two transmembrane helical regions
(TM1 and TM2) divide CPT1A protein into the small N-terminal regulatory region (about 47 amnio acids) and the main C-terminal catalytic domain. (B)
Dual physiological functions of CPT1A.
Frontiers in Pharmacology frontiersin.org03
Liang 10.3389/fphar.2023.1160440
Physiological functions of CPT1A
Succinylation
Lysine succinylation is a newly identied protein post-
translational modication (Zhang et al., 2011). CPT1A is one
of four well-known succinylation regulators including CPT1A,
lysine acetyltransferase 2A (KAT2A), Sirtuin5 (SIRT5) and
Sirtuin7 (SIRT7) (Wang et al., 2017;Kurmi et al., 2018;Lu
et al., 2021). CPT1A mutant H473A (key acyl-CoA binding
site) loses all acylation activity; mutant G710E loses the
carnitine palmitoyl-transferase activity, however, maintains its
succinylation activity, suggesting that Gly710 is very important
for CPT1A succinyl-transferase activity (Kurmi et al., 2018).
CPT1A can modulate the succinylation of enolase 1 to
enhance the growth of breast cancer (Kurmi et al., 2018)and
the succinylation of S100A10 to facilitate the spread of gastric
cancer (Wang et al., 2019).
CPT1A in iTreg cell differentiation
Inducible regulatory T (iTreg) cells are critical for immune
suppression and maintains the immune homeostasis
(Josefowicz et al., 2012;Savage et al., 2013). Butyric acid can
be processed to butyryl-CoA, which competes with malonyl-
CoA at His473 to release CPT1A activity for FAO, thereby
inducing inducible regulatory T cell (iTreg cell)
differentiation via Butyric acid- Butyryl CoACPT1A axis
(Hao et al., 2021).
Regulation of CPT1A
The balance of lipid metabolism is crucial for maintaining
homeostasis, making it crucial to control CPT1A, the key enzyme
of FAO. Genetic, physiological, and dietary modulators are all
involved in the regulation of CPT1A (Figure 5).
Transcriptional regulation of CPT1A
PPARαProliferative activated receptor α(PPARα) can greatly
enhance CPT1A expression (Kersten et al., 1999). The transcriptional
coactivator PPAR gamma coactivator 1α(PGC-1α) cooperates with
PPARαto modulate CPT1A in the liver (Jackson-Hayes et al., 2003).
Besides, the regulation of CPT1A by Leucine-rich repeat kinase 2
(LRRK2) may be through the activation of AMP-activated protein
kinase (AMPK) and PPARα(Lin et al., 2020).
FGF21 The PPARα-broblast growth factor 21 (FGF21) axis
was activated in the liver of CPT1A decient mice, which could be
used to avoid inammation, insulin resistance, and weight gain
(Sun et al., 2021). Besides, FGF21 could promote CPT1A
expression and FAO in β-cells by activating the AMPK-ACC
(acetyl-CoA carboxylase) pathway and PPAR δ/γsignaling axis
(Xie et al., 2019).
PGC-1αPGC1αbinds to CCAAT/enhancer binding protein β
(CEBPB) to enhance CPT1A transcription, resulting in activation of
FAO through PGC1α/CEBPB/CPT1A/FAO signaling axis, which
can promote radiation resistance of nasopharyngeal carcinoma
(NPC) (Du et al., 2019).
MiRNA regulation of CPT1A
MiR-33a/b Several genes involved in fatty acid metabolism
including CPT1A, HADHB contain predicted binding sites for
miR-33a/b (Davalos et al., 2011). Overexpression of miR-33a/b
can reduce FAO by downregulating CPT1A and lead to the
accumulation of triglycerides in human hepatic cells (Davalos
et al., 2011).
MiR-124 Downregulation of CPT1A expression by miR-124,
limiting the conversion of long-chain acyl-CoA moieties to long-
chain acylcarnitine (Valentino et al., 2017).
MiR-328-3p CPT1A is a downstream target of miR-328-3p in
breast cancer, and miR-328-3p overexpression suppresses cancer
spread by interfering with FAO via CPT1A (Zeng et al., 2022). The
MiR-328-3p-CPT1A-FAO pathway is crucial for the metastasis of
FIGURE 4
CPT1A homo-trimer structure predicted by ClusPro online server (https://cluspro.org/). (A) Top view of CPT1A trimer; (B) Bottom view of CPT1A
trimer with transmembrane region highlighted by yellow circle; (C) Side view of CPT1A trimer located on mitochondrial outer membrane.
Frontiers in Pharmacology frontiersin.org04
Liang 10.3389/fphar.2023.1160440
breast cancer, and miR-328-3p upregulation can be used for
reducing metastasis in breast cancer patients (Zeng et al., 2022).
MiR-370 The liver miR-370 plays a signicant role in the
inhibition of CPT1A gene expression, which can directly affect
its target gene CPT1A, suppressing its expression and lowering
FAO efcacy (Iliopoulos et al., 2010).
Hormone regulation of CPT1A
Hormones can affect CPT1A in organs like the heart, liver, and
muscle. Several hormones can take involvement in CPT1A
expression and enzyme activity control.
Insulin Insulin can signicantly lower the expression of CPT1A
and increase its sensitivity to malonyl-coA (Park et al., 1995). By
controlling CPT1A expression and enzyme activity, insulin can limit
FAO and gluconeogenesis in order to lower blood sugar level, which
is also one of the mechanisms to treat diabetes.
Thyroxine Thyroxine, a different hormonal modulator, has a
signicant impact on FAO in the liver. CPT1A mRNA could rise by
ve times with thyroxine supplementation, but mRNA in
hypothyroid rats decreased (Heimberg et al., 1985).Thyroxine can
increase CPT1A expression by interacting with the thyroid response
elements (TRE) in the CPT1A promoter, (Mynatt et al., 1994).
Thyroxine can also assist in the upregulation of the CPT1A gene by
increasing PGC-1αmRNA and protein levels in hepatocytes (Zhang
et al., 2004).
ERRαEstrogen-Related Receptor-α(ERRα), which belongs to
the nuclear receptor subfamily, is a viable target for NAFLD and that
the ERRαagonist may serve as an intriguing pharmacological option
for management of metabolic diseases (Mao et al., 2022). ERRα
reduces the thyroid hormone-induced expression of CPT1A and
mitochondrial FAO via PGC1α(Singh et al., 2018). Inhibition of
ERRαwith XCT790 treatment can increase the expression of
CPT1A, further promoting lipid metabolism (Michalek et al., 2011).
Adiponectin Adiponectin, an adipose-secreted protein that has
been linked to insulin sensitivity, plasma lipids, and inammatory
patterns, is an established biomarker for metabolic health
(Aslibekyan et al., 2017). Adiponectin phosphorylates and
triggers AMPK, which modulates CPT1A via the AMPK-ACC-
CPT1A pathway (Li et al., 2007). The acetyl-CoA carboxylase
(ACC)/malonyl-CoA pathway could be strongly blocked by
phosphorylated AMPK, thus increasing the activity of the CPT1A
enzyme. Besides, CPT1A methylation is associated with circulating
adiponectin levels, likely in an obesity-dependent manner, which
can be a novel pleiotropic marker of chronic disease risk (Aslibekyan
et al., 2017).
Metabolite regulation of CPT1A
Long chain fatty acid (LCFA) LCFA is a substrate for CPT1A and
one natural ligand of PPARα(Nakamura et al., 2014). It can up-
regulate CPT1A by directly acting on peroxisome proliferator
response elements (PPREs) in CPT1A introns, as well as via
activating PPARα(Chatelain et al., 1996;Le May et al., 2005).
Malonyl-CoA Malonyl-CoA is produced by acetyl-CoA
carboxylase2 (ACC2) during fatty acid synthesis, and is a natural
inhibitor of CPT1A. CPT1A is sensitive to malonyl-CoA, and the
sensitivity depends on the concentration of malonyl-CoA (Robinson
and Zammit, 1982). However, fasting or insulin decit signicantly
reduces CPT1As sensitivity to malonyl-CoA (Park et al., 1995;
Akkaoui et al., 2009). CPT1A structure has a short N-terminus and a
major C-terminus containing a catalytic site and a malonyl-CoA
binding site. Malonyl-CoA inhibition of CPT1A will be lost as a
result of the specic area of the N-terminal interacting with the
FIGURE 5
Regulation of CPT1A at protein level (enzyme activity) (A) and gene expression level (B).
Frontiers in Pharmacology frontiersin.org05
Liang 10.3389/fphar.2023.1160440
malonyl-CoA binding site in the C-terminal (Morillas et al., 2004;
Lopez-Vinas et al., 2007;Rufer et al., 2009;Rao et al., 2011).
Malonyl-CoA inhibits CPT1A activity through allosteric
inhibition and in a concentration-dependent manner, and the
C-termini of CPT1A served as the malonyl-CoA binding site
(Rao et al., 2011). The afnity for malonyl-CoA can also be
inuenced by the N-terminal state of CPT1A (residues 142),
which has two possible conformational states (Nαinhibitory state
and Nβnon-inhibitory state) (Lopez-Vinas et al., 2007;Rao et al.,
2011). The presence of a curved, amphiphilic binding surface is
necessary for the non-inhibitory state (Rao et al., 2011).
CPT1A activity can be regulated via the AMPK-ACC-CPT1A
axis (Figure 6)(Yao et al., 2020). ACC is inhibited through
phosphorylation by AMPK, and ACCs inhibiting effect on
CPT1A is relieved by lowering the malonyl-CoA level (Yao et al.,
2020).
CPT1A in diseases
CPT1A deciency
CPT1A deciency is a rare mitochondrial FAO disorder caused
by autosomal recessive mutations (Bellusci et al., 2017). The CPT1A
deciency can trigger a variety of illnesses, including hepatic
encephalopathy, recurrent hypoglycemia, hepatomegaly,
hyperammonemia, renal tubular acidosis, and so on (Collins
et al., 2010). Rapid onset, frequent recurrence, and high mortality
are signicant features for CPT1A deciency. Symptoms are
classied into three classes according to the frequency (Figure 7).
Mutations associated with CPT1A deciency can be divided into
two categories: type one affect directly the catalytic center, which
results in loss of activity (functional determinant); type two affects
the stability of the enzyme, which indirectly decreases the catalytic
efciency (structural determinant) (Gobin et al., 2003).
Mutations A275T, R357W, A414V, L484P, and Y498C, far away
from the active region, decrease activity by affecting the stability of
CPT1A (Gobin et al., 2003). C304W, P479L, and R395 deletion
almost lose activity (Brown et al., 2001). High mortality rate mutants
P479L is a frequent CPT1A mutation found in Arctic regions like
Canada (Rajakumar et al., 2009;Gessner et al., 2010). Additionally,
G709E and G710E, two sites essential for the hydrophobic core in
the catalytic site, can abolish CPT1A activity (Dai et al., 2000). The
hydrophobic catalytic core is altered when Gly709 or Gly710 is
changed to a Glu, which introduces a large and negatively charged
group (Gobin et al., 2003). This mutation is close to the catalytic
His473 residue and the carnitine binding site, which can
signicantly change the hydrophobic pocket of CPT1A (Gobin
et al., 2003). We compiled the majority of the reported natural
mutations so far and mapped them onto CPT1A structure model
(Figure 8)(Table 1).
Currently, there is no specic drug for CPT1A deciency, and
diet control is the main therapy. Tubular acidosis can be improved
after treatment with medium chain triglyceride in CPT1A deciency
patients (Falik-Borenstein et al., 1992). Therefore, timely screening
and diagnosis of CPT1A defects appear to be particularly important,
and CPT1A defects can be nally conrmed based on results from
multiple aspects (Figure 7).
CPT1A and metabolic diseases
Patients with metabolic syndrome, obesity, and type 2 diabetes
frequently have non-alcoholic fatty liver disease,
hypertriglyceridemia, and other lipid metabolism abnormalities,
which can be somewhat improved by increasing CPT1A
expression (Deprince et al., 2020). Increased CPT1A expression
has been shown to drastically lower liver triglyceride levels
(Stefanovic-Racic et al., 2008), and suppress JNK factor to
prevent the inammatory response brought on by free fatty acids
(Gao et al., 2011). Fatty acid accumulation can cause the
development of insulin resistance, which can eventually result in
type 2 diabetes and hyperinsulinemia (Levin et al., 2007; Pan et al.,
1997).
CPT1A in vascular diseases
Metabolism of endothelial cell depends on FAO to resist
oxidative stress in the development of blood vessels, and CPT1A
plays a crucial role in this process, offering a new potential target for
the treatment of vascular-related diseases (Rohlenova et al., 2018).
CPT1A in heart failure
Heart failure is one of the top causes of death and disability
around the world, but there is yet no safe and effective clinical
therapy for heart failure (Bui et al., 2011). The clinical study of the
CPT1A inhibitor etomoxir showed improvement in heart failure,
but was ended prematurely due to elevated liver transaminase in
enrolled patients (Holubarsch et al., 2007).
Perhexiline was an effective anti-angina drug used in the last
century, but was recalled by the manufacturer because of
hepatotoxicity and peripheral neurotoxicity (Ashraan et al.,
2007). Later studies conrmed that Perhexiline could selectively
block CPT1A in liver and heart, and had an ideal inhibitory effect on
FAO and improved the oxidation of carbohydrate in the heart
(Ashraan et al., 2007).
CPT1A in NAFLD
The global obesity epidemic has dramatically increased the
prevalence of non-alcoholic fatty liver disease (NAFLD), with no
FIGURE 6
Summary of CPT1A-related diseases.
Frontiers in Pharmacology frontiersin.org06
Liang 10.3389/fphar.2023.1160440
approved treatment now (Weber et al., 2020). Decient CPT1A
expression in the liver results in a healthy steatotic state that protects
against high-fat diet-induced liver damage and increases adipose
browning in a PPARα-FGF21 axis dependent manner, suggesting
that inhibition of hepatic CPT1A may serve as a viable strategy for
the treatment of obesity and NAFLD (Weber et al., 2020;Sun et al.,
2021).
CPT1A in multiple sclerosis
Inuits have a low prevalence of multiple sclerosis, possibly
associated with in CPT1A P479L mutation (Morkholt et al.,
2019). This point mutation result in 22% residual activity of the
CPT1A (Collins et al., 2010). CPT1A inhibition may represent a
prospective therapeutic therapy for multiple sclerosis.
CPT1A in renal brosis
Renal brosis is a result of several types of chronic kidney
diseases, and currently, the only treatment is to control blood
pressure and blood sugar levels (Drawz and Rosenberg, 2013).
With conditional overexpression of CPT1A, mitochondrial
dysfunction in the brosis kidney can be alleviated, and renal
brosis can be signicantly decreased (Miguel et al., 2021). Gain
of function in CPT1A strategy may be a novel approach to treating
brosis in renal brosis (Miguel et al., 2021).
CPT1A and cancer
Oxidative stress is the key element that causes prostate cancer to
develop (Khandrika et al., 2009). Androgens may raise the levels of
CPT1A and the accumulation of reactive oxygen species, which are
closely linked to prostate cancer cell proliferation and differentiation
(Joshi et al., 2020). Currently, the primary treatments for prostate
cancer include diet modication and the use of antioxidants, while
CPT1A inhibition may provide novel therapeutic options (Lin et al.,
2010).
One characteristic of cancer is metabolic reprogramming, which
provides tumor cells the basic elements for fast cell growth and
maintains cell survival under hypoxic or nutrient-decient
conditions (Yoshida, 2015). Abnormal fat metabolism has a
profound impact on cell carcinogenesis. As an important source
of NADH, NADPH, FADH2 and ATP, FAO plays a key role in
various stages of tumor occurrence, development and metastasis
(Wang et al., 2021). CPT1A is the critical rate-limiting enzyme in
FAO, responsible for transporting fatty acids from cytoplasm to
mitochondria for oxidation. There is evidence that CPT1A is crucial
for metabolic adaptability in the development of cancer, and CPT1A
inhibition slows the spread of cancer (Tang et al., 2022).
FIGURE 7
On the left are the symptoms of CPT1A deciency gathered from Genetic and Rare Diseases (GARD) Information Center (https://rarediseases.info.
nih.gov/). On the right is the conrmatory screening process for CPT1A deciency diagnosis. The normal value of the C0/(C16 +C18) ratio is 278.75; Total
carnitine is normal at 3370 mmol/L and free carnitine is normal at 2852 mmol/L (Dowsett et al., 2017).
FIGURE 8
CPT1A deciency related mutations reported in patients. (A) The
CPT1A structure (from AlphaFold database) is colored by grey. The Cα
atoms of the CPT1A mutations in patients are shown as red spheres.
R123C, C304W, T314I, R316G, F343V, R357W, E360G, R395
(missing), A414V, D454G, G465W, P479L, L484P, Y498C, G709E, and
G710E are among the missense mutations discovered in CPT1A.
Frontiers in Pharmacology frontiersin.org07
Liang 10.3389/fphar.2023.1160440
According to growing amounts of experimental evidence
published in reputable journals in recent years, CPT1A may be
a signicant drug target for a number of cancer cells, including
breast cancer (Xiong et al., 2018;Tan et al., 2021), prostate cancer
(Schlaepfer et al., 2014;Joshi et al., 2019), glioblastoma (Jiang et al.,
2022;Kim et al., 2022;Luo et al., 2022), colon cancer (Wang et al.,
2018), gastric cancer (Wang et al., 2019;Wang et al., 2020),
multiple myeloma (Shi et al., 2016), nasopharyngeal cancer
(Tan et al., 2018;Tang et al., 2022), etc. CPT1 plays an
important role in the occurrence and development for these
cancers, and pharmacological inhibition of CPT1A can
effectively inhibit cancer cell proliferation, which makes CPT1 a
possible molecular marker for tumor diagnosis and a new target for
anti-tumor therapy.
CPT1A in breast cancer
Breast cancer is the most prevalent and leading cause of cancer
death among women globally, which has a poor prognosis, a high
rate of recurrence and metastasis, and a high fatality rate (Waks and
Winer, 2019). In 2020, there were 2.26 million new instances of
breast cancer worldwide, overtaking lung cancer (2.21 million) for
the rst time to take the top place among all cancers, according to
the International Agency for Research on Cancers (IARC) estimate
of the global cancer (Ferlay et al., 2021).
Invasion and lymphangiogenesis in breast cancer cells can be
inhibited by CPT1A knockdown, and CPT1A-null Human Dermal
Lymphatic Endothelial Cells (HDLEC cells) consistently showed
impaired invasion and lymphangiogenesis (Xiong et al., 2018).
CPT1A knockdown reduced the expression of lymphangiogenic
markers like Vascular endothelial growth factor receptor-3
(VEGFR-3) in HDLEC cells via acetyl-CoA-mediated Histone
H3 lysine 9 acetylation (H3K9ac), which can be reversed by the
addition of acetate (Xiong et al., 2018).
Myc-overexpressing triple-negative breast cancer (TNBC) has a
greater bioenergetic dependence on FAO, and CPT1A can be
pharmaceutically inhibited to reduce energy metabolism in Myc-
overexpressing TNBC cells and stop tumor growth in a xenograft
model of Myc-overexpressing TNBC (Camarda et al., 2016;Park et al.,
2016). This suggests that CPT1A inhibition may be a promising
therapeutic approach for this particular subtype of breast cancer.
Besides, Chemotherapy (CT) and radiotherapy (RT) target
rapidly dividing cells but still have signicant normal tissue
toxicity. One indicator of RT and CT resistant tumor cells is
thought to be the abnormal upregulation of CPT1A-dependent
FAO (Corn et al., 2020). High CPT1A expression, increased
FAO, and a poor prognosis are characteristics of radiation-
resistant breast cancer cells (Corn et al., 2020). Radiation
resistant breast cancer cells respond to ionizing radiation by
increasing FAO and ATP production, resulting in increased
phosphorylation of extracellular signal regulated kinase 1/2
(ERK1/2), decreased apoptosis, and promotes a more aggressive
phenotype (Han et al., 2019). Drug candidates such as Etomoxir or
its analogs, which inhibits CPT1A and FAO, can be developed as RT
and CT sensitizers in breast cancer (Han et al., 2019).
CPT1A in prostate cancer
The incidence of prostate cancer ranks the second among male
malignant tumors in the world (Schlaepfer et al., 2014). In
United States, prostate cancer has surpassed lung cancer,
becoming the most serious malignant tumor (Carlsson et al.,
2012). China has lower rates of prostate cancer than Western
nations, but these rates have been rising recently (Culp et al., 2020).
TABLE 1 Natural mutations of CPT1A found in patients.
Gene Protein Disease Description Effect References
c.367C>T R123C CPT1A deciency Brown et al. (2001)
c.912C>T/c.912C>G C304W CPT1A deciency decreased stability Brown et al. (2001)
c.941C>T T314I CPT1A deciency Stoler et al. (2004)
c.946C>G R316G CPT1A deciency Bennett et al. (2004)
c.1027T>G F343V CPT1A deciency Bennett et al. (2004)
c.1069C>T R357W CPT1A deciency decreased stability Brown et al. (2001)
c.1079A>G E360G CPT1A deciency reduced protein levels Ogawa et al. (2002)
R395 (missing) CPT1A deciency loss of activity Brown et al. (2001)
c.1241C>T A414V CPT1A deciency decreased activity Gobin et al. (2002), Gobin et al. (2003)
c.1361A>G D454G CPT1A deciency loss of activity IJlst et al. (1998)
c.1393G>T G465W CPT1A deciency Bennett et al. (2004)
c.1436C>T P479L CPT1A deciency decreased activity Brown et al. (2001)
c.1451T>C L484P CPT1A deciency decreased stability Brown et al. (2001)
c.1493A>G Y498C CPT1A deciency decreased activity Gobin et al. (2002), Gobin et al. (2003)
c.2126G>A G709E CPT1A deciency loss of activity Gobin et al. (2003)
c.2129G>A G710E CPT1A deciency loss of activity Prip-Buus et al. (2001), Gobin et al. (2003)
Frontiers in Pharmacology frontiersin.org08
Liang 10.3389/fphar.2023.1160440
Rather than using aerobic glycolysis, prostate cancer prefers
lipid for fuel. Prostate cancer cells may have less vitality after therapy
with etomoxir, irreversible inhibitor of CPT1A, and etomoxir
treatment in mice reduced xenograft growth for a period of
21 days (Schlaepfer et al., 2014). Reduced mTOR signaling,
elevated caspase-3 activation, and decreased androgen receptor
expression are linked to these outcomes (Schlaepfer et al., 2014).
The growth of prostate cancer may be aided by a stress state caused
by reactive oxygen species (ROS) that is linked to CPT1A
overexpression (Joshi et al., 2020). Besides, increased histone
acetylation has been observed in prostate cancer cells that over-
express CPT1A, suggesting that acetylation may be a means by
which CPT1A controls prostate cancer cell proliferation (Joshi et al.,
2019). These facts highlight the therapeutic potential of CPT1A
blockade to prevent prostate cancer.
CPT1A in lung cancer
Lung cancer is one of the most common malignant tumors in
humans, and its incidence and mortality are increasing year by year
worldwide (de Sousa and Carvalho, 2018). Cisplatin is one widely
used chemotherapy drugs for lung cancer (OByrne et al., 2011;
Wang and Lippard, 2005). Knockdown of CPT1A can promote
tumor cell susceptibility to Cisplatin. The CPT1A inhibitor etomoxir
can affect and coordinate with the conventional chemotherapy drug
cisplatin to increase tumor cell sensitivity to the chemotherapeutic
agent, inhibit tumor cell proliferation and promote apoptosis, thus
providing a novel approach to improving the efcacy of
chemotherapy in non-small cell lung cancer (Dheeraj et al., 2018;
Hoy et al., 2021).
CPT1A in glioblastoma multiforme (GBM)
GBM is the most common and difcult central nervous system
malignancies, with a 5-year survival rate of 6.8% (Ostrom et al.,
2020). Radiotherapy is the primary treatment for GBM, and
radiotherapy plus immunotherapy is emerging as a new option
due to the strong resistance and poor efcacy of GBM to
radiotherapy (Arina et al., 2020). However, tumor cells after
radiotherapy develop a tolerance to immunotherapy, allowing
tumor cells to escape the killing of immune cells, leading to
treatment failure (Darragh et al., 2018). After treatment of radio-
resistant GBM cells with the CPT1A inhibitor etomoxir, Oxygen
Consumption Rate (OCR) and ATP production were signicantly
inhibited, suggesting that energy conversion from glycolysis to FAO
occurs in radio-resistant GBM cells (Jiang et al., 2022). CPT1A and
CD47 are highly expressed in radiotherapy resistant GBM tumors,
and inhibition of CPT1A can result in decreased CD47 expression
and increased macrophage phagocytosis of tumor cells (Jiang et al.,
2022). High expression of CPT1A not only enhances radiotherapy
resistance in GBM tumor cells, but also enhances immune escape of
macrophages through CD47, suggesting CPT1A as a novel strategy
for the treatment of recurrent GBM multiforme.
CPT1A in colon cancer
Colon cancer develops in adipose-rich microenvironment, and
CPT1A overexpression is crucial for adipocytes to promote tumor
growth in colon cancer (Wen et al., 2017;Pearce et al., 2018). CPT1A
upregulation is a key metabolic alteration that cancer cells use to
promote β-catenin acetylation and activation, while knockdown of
CPT1A can reduce the expression of genes linked with colon cancer
cells downstream of Wnt/β-catenin (Wen et al., 2017). Overall,
CPT1A inhibition may be a useful strategy to lessen the promotion
impact of adipocytes on colon cancer.
CPT1A in gastric cancer
Gastric cancer is a malignancy of stomach lining (Torre et al.,
2015;Sung et al., 2021). In gastric cancer, the calcium-binding
cytosolic protein S100A10 is overexpressed and is essential for the
invasion and migration of tumor cells (El-Rifai et al., 2002).
CPT1A succinylated S100A10 at lysine 47, and the degree of
succinylation was elevated in human gastric cancer (Wang
et al., 2019). In summary, S100A10 succinylation promotes
gastric cancer progression and is regulated by CPT1A-mediated
succinylation and sirtuin5 (SIRT5)-mediated desuccinylation
(Wang et al., 2019).
CPT1A in ovarian cancer
CPT1A was found to be highly expressed in ovarian cancer, and
its overexpression is linked to a poor survival in ovarian cancer
patients (Shao et al., 2016;Tan et al., 2018). CPT1A inactivation
reduced cellular ATP levels and caused cell cycle arrest at G0/
G1 stage, implying that ovarian cancer cells rely on CPT1A-
mediated FAO for cell cycle progression (Shao et al., 2016).
CPT1A in nasopharyngeal carcinoma
Nasopharyngeal carcinoma (NPC) incidence can be affected by
genetic susceptibility, and environmental factors. In NPC cells,
CPT1A was the only up-expressed carnitine palmitoyl transferase
(Tang et al., 2022). Upregulated CPT1A enhances the production of
nucleoside metabolic intermediates that promote cell cycle
progression is increased in NPC cells (Tang et al., 2022). Belgian
scientists revealed via isotope labeling that palmitate-derived
carbons considerably augmented the Krebs cycle and could be
integrated into nucleotide precursors such as aspartic acid, and
pyrimidine nucleoside triphosphate (Schoors et al., 2015). Inhibiting
CPT1A causes cells to deplete stored aspartic acid and dNTP,
impairs de novo dNTP synthesis, and inhibits NPC cell cycles
DNA replication at G1/S transition, implying a potential
treatment strategy for NPC based on lipid metabolism regulation
(Schoors et al., 2015;Tan et al., 2018).
Furthermore, radiation resistance is still a signicant barrier for
NPC treatment (Chua et al., 2016;Tan et al., 2018). NPC radiation
resistance may be enhanced via the PPAR coactivator-1α(PGC1α)/
CCAAT/enhancer binding protein β(CEBPB)/CPT1A/FAO
signaling axis (Du et al., 2019). Radiation-resistant NPC cells
consistently displayed active up-regulation of CPT1A, and
inhibition of CPT1A could render NPC cells once again
vulnerable to radiation treatment by inducing mitochondrial
apoptosis (Tan et al., 2018).
CPT1A in leukemia
In acute myeloid leukemia (AML), overexpression of CPT1A
indicates poor clinical prognosis, and strong synergistic inhibitory
Frontiers in Pharmacology frontiersin.org09
Liang 10.3389/fphar.2023.1160440
effects on AML were seen when the CPT1A-selective inhibitor
ST1326 and the Bcl-2 inhibitor ABT199 were applied in
combination (Mao et al., 2021). Overall, CPT1A expression is
abnormally high in AML, and targeted suppression of CPT1A
has potent anti-leukemic effects, suggesting that CPT1A might be
a therapeutic target for the treatment of AML (Mao et al., 2021).
CPT1A as a target for cancer treatment
CPT1A inhibitors can lessen the survivability of cancer cells, so
CPT1A may be a useful target for cancer therapy. The main
drawback of CPT1A blockage is the undesirable impact on non-
tumor cells given the extensive tissue distribution of CPT1A.
Unfortunately, there has not yet been any evidence of apparent
selectivity against other CPT1 isoforms when developing small
molecules as CPT1A inhibitors.
CPT1A and drug development
CPT1A is an intriguing target with signicant potential for
pharmacological application. For decades, drugs targeting CPT1A
have been the focus of research on diseases like type 2 diabetes
(T2D), obesity, and other disorders (Rufer et al., 2009). Therefore,
drug development targeting CPT1A has attracted much attention.
CPT1A inhibitors
The efforts to study the molecular mechanisms of CPT1A
inhibition in disease intervention have increased in recent years due
to the association with cancer (Schlaepfer et al., 2014). However, there
are few small-molecule inhibitors of CPT1A. In the past, efforts were
concentrated on CPT1A inhibitors, primarily two main kinds of
inhibitors including substrate derivatives, glycidic acid derivatives,
and malonyl-CoA analogues (Figure 9)(Ceccarelli et al., 2011).
Etomoxir and 2-tetradecylglycidic acid (TDGA) are both
ethylene oxide compounds. Etomoxir and TDGA can bind to the
active site of CPT1A to produce inhibitory effects (Brady and Brady,
1986;Ratheiser et al., 1991). These two inhibitors belong to the rst
developed class. However, Etomoxir lacks selectivity to
CPT1 isoforms, and TDGA can affect the renin angiotensin
system, resulting in myocardial hypertrophy and other side
effects (Brady and Brady, 1986). Etomoxir is a strong irreversible
inhibitor of CPT1A (Selby and Sherratt, 1989). However, preclinical
research was stopped because of hepatotoxicity after a phase II
clinical trial (Holubarsch et al., 2007).
An amino-carnitine analog called ST1326 (oral formulation-
Teglicar) was initially created for diabetic ketoacidosis (Giannessi
et al., 2003). ST1326 belongs to formyl-carnitine derivatives, which
has a specic and reversible inhibitory effect on liver CPT1A, and is
currently used as a novel anti-hyperglycemic drug (Conti et al.,
2011). Compared to etomoxir, ST1326 (oral formulation-Teglicar) is
more selective for CPT1A while CPT1B can also be inhibited by
etomoxir (Conti et al., 2011). ST1326 can signicantly improve
hyperglycemia and adjust the dynamic balance of glucose in obesity
and type 2 diabetes models, showing good application prospects
(Conti et al., 2011).
It was discovered that C75 directly stimulated CPT1A activity
(Idrovo et al., 2012). However, it was also found that CPT1A is
inhibited by low amounts of C75 that have been converted to C75-
CoA (Mera et al., 2009). There is still debate about this matter,
though.
Perhexiline, a clinical CPT1A/CPT2 inhibitor, is approved for
the management of angina pectoris outside of the United States
(Ashraan et al., 2007). Perhexiline increases the efciency of
oxygen consumption by blocking FAO and switching the energy
metabolism from lipid to carbohydrate. Despite being effective for
angina pectoris, perhexiline was reported with neurotoxicity and
hepatotoxicity for prolonged use (Ren et al., 2020).
CPT1A is a signicant therapeutic target due to its important
function in a variety of illnesses (Schlaepfer and Joshi, 2020).
CPT1As protein structure is still unknown, though. It is crucial
to discover the structure and catalytic role of the enzyme in order to
design more powerful inhibitors. To avoid side effects, selectivity
needs to be considered further. For instance, the fact that perhexiline
targets both CPT1A and CPT2 raises the possibility that the negative
effects may be related to its lack of isoform selectivity.
The isoform selectivity of CPT1A inhibitor has a signicant
impact on how the therapeutic benet is evaluated. The best-studied
CPT1 inhibitor, etomoxir, may have the problem of off-target side
effect (Yao et al., 2018). When assessing safety, liver selectivity
should also be taken into account for CPT1A.
CPT1A agonists
Moderately increasing the expression or activity of CPT1A can
promote FAO and improve a variety of metabolic diseases caused by
high fat diet (Figure 10).
C75 and baicalin can directly activate CPT1A (Idrovo et al.,
2012;Fang et al., 2020). In addition, several small molecules can
indirectly activate CPT1A by acting on ACC2, including the AMPK
activators metformin and AICAR (Tomita et al., 2005), and the
ACC2 inhibitors TOFA and Soraphen (Agostini et al., 2022). Anti-
Sense Oligonucleotide (ASO)-mediated inhibition of ACC2 may
also play a role in activating CPT1A. CPT1A can also be indirectly
promoted by PPAR activators Fibrates and GW501516 (Cox, 2017).
However, overexpression of CPT1A may increase the FAO rate,
promote cell metabolism, and accelerate cell apoptosis, which brings
certain difculties for the development of related drugs.
FIGURE 9
Four well-studied inhibitors (Etomoxir, Perhexiline, ST1326,
TDGA) of CPT1A.
Frontiers in Pharmacology frontiersin.org10
Liang 10.3389/fphar.2023.1160440
Conclusion and perspectives
In summary, as a key enzyme in FAO, CPT1A affects the
occurrence and development of a variety of diseases. The
physiological function and impact of CPT1A are becoming
increasingly better understood, which has important guiding
signicance for the research of CPT1A related diseases, and
also provides support for the drug development and application
based on targeting CPT1A. CPT1A has become a focus of
pharmaceutical research due to CPT1A pathogenic mutations
and abnormal expression in malignancies. Interest in CPT1As
role in cancer has increased recently. The mechanisms
through which CPT1A aids in cancer cell survival remain not
fully claried. Given that CPT1A was reported to promote
anoikis-resistance and metastasis in cancers like colorectal cancer,
CPT1A would be a desirable target to counteract resistance of
anticancer drugs (Wang et al., 2018). Although currently
available small-molecule drugs targeting CPT1A have shown
promising therapeutic effects, their off-target effects and
side effects are still the biggest obstacles to their application.
Finally, further research on the structure of CPT1A will
continue to improve the specicity of drug selectivity to
CPT1A to avoid off-target effect and other undesired side effects,
which will provide safer and more effective drugs for the clinical
therapy.
Author contributions
KL wrote the manuscript, made the gures, and approved the
nal manuscript.
Funding
This work is nancially supported by the Boya Postdoctoral program.
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated organizations,
or those of the publisher, the editors and the reviewers. Any product
that may be evaluated in this article, or claim that may be made by its
manufacturer, is not guaranteed or endorsed by the publisher.
References
Agostini, M., Melino, G., Habeb, B., Calandria, J. M., and Bazan, N. G. (2022).
Targeting lipid metabolism in cancer: Neuroblastoma. Cancer Metastasis Rev. 41,
255260. doi:10.1007/s10555-022-10040-8
Akkaoui, M., Cohen, I., Esnous, C., Lenoir, V., Sournac, M., Girard, J., et al. (2009).
Modulation of the hepatic malonyl-CoA-carnitine palmitoyltransferase 1A partnership
creates a metabolic switch allowing oxidation of de novo fatty acids. Biochem. J. 420,
429438. doi:10.1042/BJ20081932
Arina, A., Gutiontov, S. I., and Weichselbaum, R. R. (2020). Radiotherapy and
immunotherapy for cancer: From "systemic" to "multisite. Clin. Cancer Res. 26,
27772782. doi:10.1158/1078-0432.CCR-19-2034
FIGURE 10
Strategies to directly or indirectly stimulate CPT1A. ASO, antisense oligonucleotide; PPAR, peroxisome proliferator activated receptor; ACC, acetyl-
CoA carboxylase; AMPK, adenosine monophosphate-activated protein kinase.
Frontiers in Pharmacology frontiersin.org11
Liang 10.3389/fphar.2023.1160440
Ashraan, H., Horowitz, J. D., and Frenneaux, M. P. (2007). Perhexiline. Cardiovasc
Drug Rev. 25, 7697. doi:10.1111/j.1527-3466.2007.00006.x
Aslibekyan, S., Do, A. N., Xu, H., Li, S., Irvin, M. R., Zhi, D., et al. (2017). CPT1A
methylation is associated with plasma adiponectin. Nutr. Metab. Cardiovasc Dis. 27,
225233. doi:10.1016/j.numecd.2016.11.004
Bellusci, M., Quijada-Fraile, P., Barrio-Carreras, D., Martin-Hernandez, E., Garcia-
Silva, M., Merinero, B., et al. (2017). Carnitine palmitoyltransferase 1A deciency:
Abnormal muscle biopsy ndings in a child presenting with reyes syndrome. J. Inherit.
Metab. Dis. 40, 751752. doi:10.1007/s10545-017-0041-7
Bennett, M. J., Boriack, R. L., Narayan, S., Rutledge, S. L., and Raff, M. L. (2004). Novel
mutations in CPT 1A dene molecular heterogeneity of hepatic carnitine
palmitoyltransferase I deciency. Mol. Genet. Metab. 82, 5963. doi:10.1016/j.
ymgme.2004.02.004
Bergers, G., and Fendt, S. M. (2021). The metabolism of cancer cells during metastasis.
Nat. Rev. Cancer 21, 162180. doi:10.1038/s41568-020-00320-2
Bluher, M. (2019). Obesity: Global epidemiology and pathogenesis. Nat. Rev.
Endocrinol. 15, 288298. doi:10.1038/s41574-019-0176-8
Bonnefont, J. P., Djouadi, F., Prip-Buus, C., Gobin, S., Munnich, A., and Bastin, J.
(2004). Carnitine palmitoyltransferases 1 and 2: Biochemical, molecular and medical
aspects. Mol. Asp. Med. 25, 495520. doi:10.1016/j.mam.2004.06.004
Brady, P. S., and Brady, L. J. (1986). Action in vivo and in vitro of 2-tetradecylglycidic
acid, 2-tetradecylglycidyl-CoA and 2-tetradecylglycidylcarnitine on hepatic carnitine
palmitoyltransferase. Biochem. J. 238, 801809. doi:10.1042/bj2380801
Brown, N. F., Mullur, R. S., Subramanian, I., Esser, V., Bennett, M. J., Saudubray, J.M.,
et al. (2001). Molecular characterization of L-CPT I deciency in six patients: Insights
into function of the native enzyme. J. Lipid Res. 42, 11341142. doi:10.1016/s0022-
2275(20)31604-7
Bui, A. L., Horwich, T. B., and Fonarow, G. C. (2011). Epidemiology and risk prole of
heart failure. Nat. Rev. Cardiol. 8, 3041. doi:10.1038/nrcardio.2010.165
Camarda, R., Zhou, A. Y., Kohnz, R. A., Balakrishnan, S., Mahieu, C., Anderton, B.,
et al. (2016). Inhibition of fatty acid oxidation as a therapy for MYC-overexpressing
triple-negative breast cancer. Nat. Med. 22, 427432. doi:10.1038/nm.4055
Carlsson, S., Vickers, A. J., Roobol, M., Eastham, J., Scardino, P., Lilja, H., et al. (2012).
Prostate cancer screening: Facts, statistics, and interpretation in response to the US
preventive services task force review. J. Clin. Oncol. 30, 25812584. doi:10.1200/JCO.
2011.40.4327
Ceccarelli, S. M., Chomienne, O., Gubler, M., and Arduini, A. (2011). Carnitine
palmitoyltransferase (CPT) modulators: A medicinal chemistry perspective on 35 years
of research. J. Med. Chem. 54, 31093152. doi:10.1021/jm100809g
Chatelain, F., Kohl, C., Esser, V., McGarry, J. D., Girard, J., and Pegorier, J. P. (1996).
Cyclic AMP and fatty acids increase carnitine palmitoyltransferase I gene transcription
in cultured fetal rat hepatocytes. Eur. J. Biochem. 235, 789798. doi:10.1111/j.1432-
1033.1996.00789.x
Chua, M. L. K., Wee, J. T. S., Hui, E. P., and Chan, A. T. C. (2016). Nasopharyngeal
carcinoma. Lancet 387, 10121024. doi:10.1016/S0140-6736(15)00055-0
Collins, S. A., Sinclair, G., McIntosh, S., Bamforth, F., Thompson, R., Sobol, I., et al.
(2010). Carnitine palmitoyltransferase 1A (CPT1A) P479L prevalence in live newborns
in yukon, northwest territories, and nunavut. Mol. Genet. Metab. 101, 200204. doi:10.
1016/j.ymgme.2010.07.013
Conti, R., Mannucci, E., Pessotto, P., Tassoni, E., Carminati, P., Giannessi, F., et al.
(2011). Selective reversible inhibition of liver carnitine palmitoyl-transferase 1 by
teglicar reduces gluconeogenesis and improves glucose homeostasis. Diabetes 60,
644651. doi:10.2337/db10-0346
Corn, K. C., Windham, M. A., and Rafat, M. (2020). Lipids in the tumor
microenvironment: From cancer progression to treatment. Prog. Lipid Res. 80,
101055. doi:10.1016/j.plipres.2020.101055
Cox, R. L. (2017). Rationally designed PPARδ-specic agonists and their therapeutic
potential for metabolic syndrome. Proc. Natl. Acad. Sci. U. S. A. 114, 32843285. doi:10.
1073/pnas.1702084114
Culp, M. B., Soerjomataram, I., Efstathiou, J. A., Bray, F., and Jemal, A. (2020). Recent
global patterns in prostate cancer incidence and mortality rates. Eur. Urol. 77, 3852.
doi:10.1016/j.eururo.2019.08.005
Dai, J., Zhu, H., Shi, J., and Woldegiorgis, G. (2000). Identication by mutagenesis of
conserved arginine and tryptophan residues in rat liver carnitine palmitoyltransferase I
important for catalytic activity. J. Biol. Chem. 275, 2202022024. doi:10.1074/jbc.
M002118200
Darragh, L. B., Oweida, A. J., and Karam, S. D. (2018). Overcoming resistance to
combination radiation-immunotherapy: A focus on contributing pathways within the
tumor microenvironment. Front. Immunol. 9, 3154. doi:10.3389/mmu.2018.03154
Davalos, A., Goedeke, L., Smibert, P., Ramirez, C. M., Warrier, N. P., Andreo , U., et al.
(2011). miR-33a/b contribute to the regulation of fatty acid metabolism and insulin
signaling. Proc. Natl. Acad. Sci. U. S. A. 108, 92329237. doi:10.1073/pnas.1102281108
de Sousa, V. M. L., and Carvalho, L. (2018). Heterogeneity in lung cancer.
Pathobiology 85, 96107. doi:10.1159/000487440
Deprince, A., Haas, J. T., and Staels, B. (2020). Dysregulated lipid metabolism links
NAFLD to cardiovascular disease. Mol. Metab. 42, 101092. doi:10.1016/j.molmet.2020.
101092
Dheeraj, A., Agarwal, C., Schlaepfer, I. R., Raben, D., Singh, R., Agarwal, R., et al.
(2018). A novel approach to target hypoxic cancer cells via combining beta-oxidation
inhibitor etomoxir with radiation. Hypoxia (Auckl) 6, 2333. doi:10.2147/HP.S163115
Dowsett, L., Lulis, L., Ficicioglu, C., and Cuddapah, S. (2017). Utility of genetic testing
for conrmation of abnormal newborn screening in disorders of long-chain fatty acids:
A missed case of carnitine palmitoyltransferase 1A (CPT1A) deciency. Int. J. Neonatal
Screen 3, 10. doi:10.3390/ijns3020010
Drawz, P. E., and Rosenberg, M. E. (2013). Slowing progression of chronic kidney
disease. Kidney Int. Suppl. 3, 372376. doi:10.1038/kisup.2013.80
Du, Q., Tan, Z., Shi, F., Tang, M., Xie, L., Zhao, L., et al. (2019). PGC1α/CEBPB/
CPT1A axis promotes radiation resistance of nasopharyngeal carcinoma through
activating fatty acid oxidation. Cancer Sci. 110, 20502062. doi:10.1111/cas.14011
El-Rifai, W., Moskaluk, C. A., Abdrabbo, M. K., Harper, J., Yoshida, C., Riggins, G. J.,
et al. (2002). Gastric cancers overexpress S100A calcium-binding proteins. Cancer Res.
62, 68236826.
Fado, R., Rodriguez-Rodriguez, R., and Casals, N. (2021). The return of malonyl-CoA
to the brain: Cognition and other stories. Prog. Lipid Res. 81, 101071. doi:10.1016/j.
plipres.2020.101071
Falik-Borenstein, Z. C., Jordan, S. C., Saudubray, J. M., Brivet, M., Demaugre, F.,
Edmond, J., et al. (1992). Brief report: Renal tubular acidosis in carnitine
palmitoyltransferase type 1 deciency. N. Engl. J. Med. 327, 2427. doi:10.1056/
NEJM199207023270105
Fang, P., Yu, M., Shi, M., Bo, P., Gu, X., and Zhang, Z. (2020). Baicalin and its
aglycone: A novel approach for treatment of metabolic disorders. Pharmacol. Rep. 72,
1323. doi:10.1007/s43440-019-00024-x
Faye, A., Esnous, C., Price, N. T., Onfray, M. A., Girard, J., and Prip-Buus, C. (2007).
Rat liver carnitine palmitoyltransferase 1 forms an oligomeric complex within the outer
mitochondrial membrane. J. Biol. Chem. 282, 2690826916. doi:10.1074/jbc.
M705418200
Ferlay, J., Colombet, M., Soerjomataram, I., Parkin, D. M., Pineros, M., Znaor, A.,
et al. (2021). Cancer statistics for the year 2020: An overview. Int. J. Cancer 149,
778789. doi:10.1002/ijc.33588
Fraser, F., Corstorphine, C. G., and Zammit, V. A. (1997). Topology of carnitine
palmitoyltransferase I in the mitochondrial outer membrane. Biochem. J. 323 (3),
711718. doi:10.1042/bj3230711
Gessner, B. D., Gillingham, M. B., Birch, S., Wood, T., and Koeller, D. M. (2010).
Evidence for an association between infant mortality and a carnitine
palmitoyltransferase 1A genetic variant. Pediatrics 126, 945951. doi:10.1542/peds.
2010-0687
Giannessi, F., Pessotto, P., Tassoni, E., Chiodi, P., Conti, R., De Angelis, F., et al.
(2003). Discovery of a long-chain carbamoyl aminocarnitine derivative, a reversible
carnitine palmitoyltransferase inhibitor with antiketotic and antidiabetic activity.
J. Med. Chem. 46, 303309. doi:10.1021/jm020979u
Gobin, S., Bonnefont, J. P., Prip-Buus, C., Mugnier, C., Ferrec, M., Demau gre, F., et al.
(2002). Organization of the human liver carnitine palmitoyltransferase 1 gene (CPT1A)
and identication of novel mutations in hypoketotic hypoglycaemia. Hum. Genet. 111,
179189. doi:10.1007/s00439-002-0752-0
Gobin, S., Thuillier, L., Jogl, G., Faye, A., Tong, L., Chi, M., et al. (2003). Functional
and structural basis of carnitine palmitoyltransferase 1A deciency. J. Biol. Chem. 278,
5042850434. doi:10.1074/jbc.M310130200
Han, S., Wei, R., Zhang, X., Jiang, N., Fan, M., Huang, J. H., et al. (2019). CPT1A/2-
Mediated FAO enhancement-A metabolic target in radioresistant breast cancer. Front.
Oncol. 9, 1201. doi:10.3389/fonc.2019.01201
Hao, F., Tian, M., Zhang, X., Jin, X., Jiang, Y., Sun, X., et al. (2021). Butyrate enhances
CPT1A activity to promote fatty acid oxidation and iTreg differentiation. Proc. Natl.
Acad. Sci. U. S. A. 118, e2014681118. doi:10.1073/pnas.2014681118
Heimberg, M., Olubadewo, J. O., and Wilcox, H. G. (1985). Plasma lipoproteins and
regulation of hepatic metabolism of fatty acids in altered thyroid states. Endocr. Rev. 6,
590607. doi:10.1210/edrv-6-4-590
Holubarsch,C.J.,Rohrbach,M.,Karrasch,M.,Boehm,E.,Polonski,L.,
Ponikowski, P., et al. (2007). A double-blind randomized multicentre clinical
trial to evaluate the efcacy and safety of two doses of etomoxir in comparison with
placebo in patients with moderate congestive heart failure: The ERGO (etomoxir
for the recovery of glucose oxidation) study. Clin.Sci.(Lond)113, 205212. doi:10.
1042/CS20060307
Houten, S. M., Violante, S., Ventura, F. V., and Wanders, R. J. (2016). The
biochemistry and physiology of mitochondrial fatty acid beta-oxidation and its
genetic disorders. Annu. Rev. Physiol. 78, 2344. doi:10.1146/annurev-physiol-
021115-105045
Hoy, A. J., Nagarajan, S. R., and Butler, L. M. (2021). Tumourfatty acidmetabolism in
the context of therapy resistance and obesity. Nat. Rev. Cancer 21, 753766. doi:10.1038/
s41568-021-00388-4
Frontiers in Pharmacology frontiersin.org12
Liang 10.3389/fphar.2023.1160440
Idrovo, J. P., Yang, W. L., Nicastro, J., Coppa, G. F. , and Wang, P. (2012). Stimulation
of carnitine palmitoyltransferase 1 improves renal function and attenuates tissue
damage after ischemia/reperfusion. J. Surg. Res. 177, 157164. doi:10.1016/j.jss.2012.
05.053
Ijlst, L., Mandel, H., Oostheim, W., Ruiter, J. P., Gutman, A., and Wanders, R. J.
(1998). Molecular basis of hepatic carnitine palmitoyltransferase I deciency. J. Clin.
Invest. 102, 527531. doi:10.1172/JCI2927
Iliopoulos, D., Drosatos, K., Hiyama, Y., Goldberg, I. J., and Zannis, V. I. (2010).
MicroRNA-370 controls the expression of microRNA-122 and Cpt1alpha and affects
lipid metabolism. J. Lipid Res. 51, 15131523. doi:10.1194/jlr.M004812
Indiveri, C., Iacobazzi, V., Tonazzi, A., Giangregorio, N., Infantino, V., Convertini, P.,
et al. (2011). The mitochondrial carnitine/acylcarnitine carrier: Function, structure and
physiopathology. Mol. Asp. Med. 32, 223233. doi:10.1016/j.mam.2011.10.008
Jackson-Hayes, L., Song, S., Lavrentyev, E. N., Jansen, M. S., Hillgartner, F. B., Tian, L.,
et al. (2003). A thyroid hormone response unit formed between the promoter and rst
intron of the carnitine palmitoyltransferase-Ialpha gene mediates the liver-specic
induction by thyroid hormone. J. Biol. Chem. 278, 79647972. doi:10.1074/jbc.
M211062200
Jenei, Z. A., Borthwick, K., Zammit, V. A., and Dixon, A. M. (2009). Self-associatio n of
transmembrane domain 2 (TM2), but not TM1, in carnitine palmitoyltransferase 1A:
Role of GXXXG(A) motifs. J. Biol. Chem. 284, 69886997. doi:10.1074/jbc.M808487200
Jiang, N., Xie, B., Xiao, W., Fan, M., Xu, S., Duan, Y., et al. (2022). Fatty acid oxidation
fuels glioblastoma radioresistance with CD47-mediated immune evasion. Nat.
Commun. 13, 1511. doi:10.1038/s41467-022-29137-3
Josefowicz, S. Z., Lu, L. F., and Rudensky, A. Y. (2012). Regulatory T cells:
Mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531564.
doi:10.1146/annurev.immunol.25.022106.141623
Joshi, M., Kim, J., DAlessandro, A., Monk, E., Bruce, K., Elajaili, H., et al. (2020).
CPT1A over-expression increases reactive oxygen species in the mitochondria and
promotes antioxidant defenses in prostate cancer. Cancers (Basel) 12, 3431. doi:10.3390/
cancers12113431
Joshi, M., Stoykova, G. E., Salzmann-Sullivan, M., Dzieciatkowska, M., Liebman, L.
N., Deep, G., et al. (2019). CPT1A supports castration-resistant prostate cancer in
androgen-deprived conditions. Cells 8, 1115. doi:10.3390/cells8101115
Kersten, S., Seydoux, J., Peters, J. M., Gonzalez, F. J., Desvergne, B., and Wahli, W.
(1999). Peroxisome proliferator-activated receptor alpha mediates the adaptive
response to fasting. J. Clin. Invest. 103, 14891498. doi:10.1172/JCI6223
Khandrika, L., Kumar, B., Koul, S., Maroni, P., and Koul, H. K. (2009). Oxidative
stress in prostate cancer. Cancer Lett. 282, 125136. doi:10.1016/j.canlet.2008.12.011
Kim, S. J., Park, S. J., Park, J., Cho, H. J., Shim, J. K., Seon, J., et al. (2022). Dual
inhibition of CPT1A and G6PD suppresses glioblastoma tumorspheres. J. Neurooncol
160, 677689. doi:10.1007/s11060-022-04189-z
Kurmi, K., Hitosugi, S., Wiese, E. K., Boakye-Agyeman, F., Gonsalves, W. I., Lou, Z.,
et al. (2018). Carnitine palmitoyltransferase 1A has a lysine succinyltransferase activity.
Cell Rep. 22, 13651373. doi:10.1016/j.celrep.2018.01.030
Le May, C., Cauzac, M., Diradourian, C., Perdereau, D., Girard, J., Burnol, A. F., et al.
(2005). Fatty acids induce L-CPT I gene expression through a PPARalpha-independent
mechanism in rat hepatoma cells. J. Nutr. 135, 23132319. doi:10.1093/jn/135.10.2313
Lee, K., Kerner, J., and Hoppel, C. L. (2011). Mitochondrial carnitine
palmitoyltransferase 1a (CPT1a) is part of an outer membrane fatty acid transfer
complex. J. Biol. Chem. 286, 2565525662. doi:10.1074/jbc.M111.228692
Li, L., Wu, L., Wang, C., Liu, L., and Zhao, Y. (2007). Adiponectin modulates carnitine
palmitoyltransferase-1 through AMPK signaling cascade in rat cardiomyocytes. Regul.
Pept. 139, 7279. doi:10.1016/j.regpep.2006.10.007
Lin, C. W., Peng, Y. J., Lin, Y. Y., Mersmann, H. J., and Ding, S. T. (2020).
LRRK2 regulates CPT1A to promote beta-oxidation in HepG2 cells. Molecules 25,
4122. doi:10.3390/molecules25184122
Lin, H., Lu, J. P., Laamme, P., Qiao, S., Shayegan, B., Bryskin, I., et al. (2010). Inter-
related in vitro effects of androgens, fatty acids and oxidative stress in prostate cancer: A
mechanistic model supporting prevention strategies. Int. J. Oncol. 37, 761766. doi:10.
3892/ijo_00000725
Lopez-Vinas, E., Bentebibel, A., Gurunathan, C., Morillas, M., de Arriaga, D., Serra,
D., et al. (2007). Denition by functional and structural analysis of two malonyl-CoA
sites in carnitine palmitoyltransferase 1A. J. Biol. Chem. 282, 1821218224. doi:10.1074/
jbc.M700885200
Lu, W., Che, X., Qu, X., Zheng, C., Yang, X., Bao, B., et al. (2021). Succinylation
regulators promote clear cell renal cell carcinoma by immune regulation and RNA N6-
methyladenosine methylation. Front. Cell Dev. Biol. 9, 622198. doi:10.3389/fcell.2021.
622198
Luo, M., Liu, Y. Q., Zhang, H., Luo, C. H., Liu, Q., Wang, W. Y., et al. (2022).
Overexpression of carnitine palmitoyltransferase 1A promotes mitochondrial fusion
and differentiation of glioblastoma stem cells. Lab. Invest. 102, 722730. doi:10.1038/
s41374-021-00724-0
Mao, L., Peng, L., Ren, X., Chu, Y., Nie, T., Lin, W., et al. (2022). Discovery of
JND003 as a new selective estrogen-related receptor alpha agonist alleviating
nonalcoholic fatty liver disease and insulin resistance. ACS Bio Med. Chem. Au 2,
282296. doi:10.1021/acsbiomedchemau.1c00050
Mao, S., Ling, Q., Pan, J., Li, F., Huang, S., Ye, W., et al. (2021). Inhibition of CPT1a as
a prognostic marker can synergistically enhance the antileukemic activity of ABT199.
J. Transl. Med. 19, 181. doi:10.1186/s12967-021-02848-9
McGarry, J. D., Leatherman, G. F., and Foster, D. W. (1978). Carnitine
palmitoyltransferase I. The site of inhibition of hepatic fatty acid oxidation by
malonyl-CoA. J. Biol. Chem. 253, 41284136. doi:10.1016/s0021-9258(17)34693-8
Mera, P., Bentebibel, A., Lopez-Vinas, E., Cordente, A. G., Gurunathan, C., Sebastian,
D., et al. (2009). C75 is converted to C75-CoA in the hypothalamus, where it inhibits
carnitine palmitoyltransferase 1 and decreases food intake and body weight. Biochem.
Pharmacol. 77, 10841095. doi:10.1016/j.bcp.2008.11.020
Michalek, R. D., Gerriets, V. A., Nichols, A. G., Inoue, M., Kazmin, D., Chang, C. Y.,
et al. (2011). Estrogen-related receptor-alpha is a metabolic regulator of effector T-cell
activation and differentiation. Proc. Natl. Acad. Sci. U. S. A. 108, 1834818353. doi:10.
1073/pnas.1108856108
Miguel, V., Tituana, J., Herrero, J. I., Herrero, L., Serra, D., Cuevas, P., et al. (2021).
Renal tubule Cpt1a overexpression protects from kidney brosis by restoring
mitochondrial homeostasis. J. Clin. Invest. 131, e140695. doi:10.1172/JCI140695
Morillas, M., Lopez-Vinas, E., Valencia, A., Serra, D., Gomez-Puertas, P., Hegardt, F.
G., et al. (2004). Structural model of carnitine palmitoyltransferase I based on the
carnitine acetyltransferase crystal. Biochem. J. 379, 777784. doi:10.1042/BJ20031373
Morkholt, A. S., Trabjerg, M. S., Oklinski, M. K. E., Bolther, L., Kroese, L. J., Pritchard,
C. E. J., et al. (2019). CPT1A plays a key role in the development and treatment of
multiple sclerosis and experimental autoimmune encephalomyelitis. Sci. Rep. 9, 13299.
doi:10.1038/s41598-019-49868-6
Munir, R., Lisec, J., Swinnen, J. V., and Zaidi, N. (2019). Lipid metabolism in cancer
cells under metabolic stress. Br. J. Cancer 120, 10901098. doi:10.1038/s41416-019-
0451-4
Mynatt, R. L., Park, E. A., Thorngate, F. E., Das, H. K., and Cook, G. A. (1994).
Changes in carnitine palmitoyltransferase-I mRNA abundance produced by
hyperthyroidism and hypothyroidism parallel changes in activity. Biochem. Biophys.
Res. Commun. 201, 932937. doi:10.1006/bbrc.1994.1791
Nakamura, M. T., Yudell, B. E., and Loor, J. J. (2014). Regulation of energy
metabolism by long-chain fatty acids. Prog. Lipid Res. 53, 124144. doi:10.1016/j.
plipres.2013.12.001
OByrne, K. J., Barr, M. P., and Gray, S. G. (2011). The role of epigenetics in resistance
to Cisplatin chemotherapy in lung cancer. Cancers (Basel) 3, 14261453. doi:10.3390/
cancers3011426
Ogawa, E., Kanazawa, M., Yamamoto, S., Ohtsuka, S., Ogawa, A., Ohtake, A., et al.
(2002). Expression analysis of two mutations in carnitine palmitoyltransferase IA
deciency. J. Hum. Genet. 47, 342347. doi:10.1007/s100380200047
Ostrom, Q. T., Patil, N., Ciof, G., Waite, K., Kruchko, C., and Barnholtz-Sloan, J. S.
(2020). CBTRUS statistical report: Primary brain and other central nervous system
tumors diagnosed in the United States in 2013-2017. Neuro Oncol. 22, iv1iv96. doi:10.
1093/neuonc/noaa200
Park, E. A., Mynatt, R. L., Cook, G. A., and Kash, K. (1995). Insulin regulates enzyme
activity, malonyl-CoA sensitivity and mRNA abundance of hepatic carnitine
palmitoyltransferase-I. Biochem. J. 310 (3), 853858. doi:10.1042/bj3100853
Park, J. H., Vithayathil, S., Kumar, S., Sung, P. L., Dobrolecki, L. E., Putluri, V., et al.
(2016). Fatty acid oxidation-driven src links mitochondrial energy reprogramming and
oncogenic properties in triple-negative breast cancer. Cell Rep. 14, 21542165. doi:10.
1016/j.celrep.2016.02.004
Pearce, O. M. T., Delaine-Smith, R. M., Maniati, E., Nichols, S., Wang, J., Bohm, S.,
et al. (2018). Deconstruction of a metastatic tumor microenvironment reveals a
common matrix response in human cancers. Cancer Discov. 8, 304319. doi:10.
1158/2159-8290.CD-17-0284
Prip-Buus, C., Thuillier, L., Abadi, N., Prasad, C., Dilling, L., Klasing, J., et al. (2001).
Molecular and enzymatic characterization of a unique carnitine palmitoyltransferase 1A
mutation in the Hutterite community. Mol. Genet. Metab. 73, 4654. doi:10.1006/
mgme.2001.3176
Rajakumar, C., Ban, M. R., Cao, H., Young, T. K., Bjerregaard, P., and Hegele, R. A.
(2009). Carnitine palmitoyltransferase IA polymorphism P479L is common in
Greenland Inuit and is associated with elevated plasma apolipoprotein A-I. J. Lipid
Res. 50, 12231228. doi:10.1194/jlr.P900001-JLR200
Ramsay, R. R., Gandour, R. D., and van der Leij, F. R. (2001). Molecular enzymology
of carnitine transfer and transport. Biochim. Biophys. Acta 1546, 2143. doi:10.1016/
s0167-4838(01)00147-9
Rao, J. N., Warren, G. Z. L., Estolt-Povedano, S., Zammit, V. A., and Ulmer, T. S.
(2011). An environment-dependent structural switch underlies the regulation of
carnitine palmitoyltransferase 1A. J. Biol. Chem. 286, 4254542554. doi:10.1074/jbc.
M111.306951
Ratheiser, K., Schneeweiss, B., Waldhausl, W., Fasching, P., Korn, A., Nowotny, P.,
et al. (1991). Inhibition by etomoxir of carnitine palmitoyltransferase I reduces hepatic
glucose production and plasma lipids in non-insulin-dependent diabetes mellitus.
Metabolism 40, 11851190. doi:10.1016/0026-0495(91)90214-h
Frontiers in Pharmacology frontiersin.org13
Liang 10.3389/fphar.2023.1160440
Ren, Z., Chen, S., Seo, J. E., Guo, X., Li, D., Ning, B., et al. (2020). Mitochondrial
dysfunction and apoptosis underlie the hepatotoxicity of perhexiline. Toxicol Vitro 69,
104987. doi:10.1016/j.tiv.2020.104987
Rinaldi, C., Schmidt, T., Situ, A. J., Johnson, J. O., Lee, P. R., Chen, K. L., et al. (2015).
Mutation in CPT1C associated with pure autosomal dominant spastic paraplegia. JAMA
Neurol. 72, 561570. doi:10.1001/jamaneurol.2014.4769
Robert, X., and Gouet, P. (2014). Deciphering key features in protein structures with
the new ENDscript server. Nucleic Acids Res. 42, W320W324. doi:10.1093/nar/gku316
Robinson, I. N., and Zammit, V. A. (1982). Sensitivity of carnitine acyltran sferase I to
malonly-CoA inhibition in isolated rat liver mitochondria is quantitatively related to
hepatic malonyl-CoA concentration in vivo.Biochem. J. 206, 177179. doi:10.1042/
bj2060177
Rohlenova, K., Veys, K., Miranda-Santos, I., De Bock, K., and Carmeliet, P. (2018).
Endothelial cell metabolism in health and disease. Trends Cell Biol. 28, 224236. doi:10.
1016/j.tcb.2017.10.010
Rufer, A. C., Thoma, R., and Hennig, M. (2009). Structural insight into function and
regulation of carnitine palmitoyltransferase. Cell Mol. Life Sci. 66, 24892501. doi:10.
1007/s00018-009-0035-1
Savage, P. A., Malchow, S., and Leventhal, D. S. (2013). Basic principles of tumor-
associated regulatory T cell biology. Trends Immunol. 34, 3340. doi:10.1016/j.it.2012.
08.005
Schlaepfer, I. R., and Joshi, M. (2020). CPT1A-mediated fat oxidation, mechanisms,
and therapeutic potential. Endocrinology 161, bqz046. doi:10.1210/endocr/bqz046
Schlaepfer, I. R., Rider, L., Rodrigues, L. U., Gijon, M. A., Pac, C. T., Romero, L., et al.
(2014). Lipid catabolism via CPT1 as a therapeutic target for prostate cancer. Mol.
Cancer Ther. 13, 23612371. doi:10.1158/1535-7163.MCT-14-0183
Schoors, S., Bruning, U., Missiaen, R., Queiroz, K. C., Borgers, G., Elia, I., et al. (2015).
Fatty acid carbon is essential for dNTP synthesis in endothelial cells. Nature 520,
192197. doi:10.1038/nature14362
Selby, P. L., and Sherratt, H. S. (1989). Substituted 2-oxiranecarboxylic acids: A new
group of candidate hypoglycaemic drugs. Trends Pharmacol. Sci. 10, 495500. doi:10.
1016/0165-6147(89)90049-7
Shao, H., Mohamed, E. M., Xu, G. G., Waters, M., Jing, K., Ma, Y., et al. (2016).
Carnitine palmitoyltransferase 1A functions to repress FoxO transcription factors to
allow cell cycle progression in ovarian cancer. Oncotarget 7, 38323846. doi:10.18632/
oncotarget.6757
Shepherd, P. R., and Kahn, B. B. (1999). Glucose transporters and insulin action--
implications for insulin resistance and diabetes mellitus. N. Engl. J. Med. 341, 248257.
doi:10.1056/NEJM199907223410406
Shi, J., Fu, H., Jia, Z., He, K., Fu, L., and Wang, W. (2016). High expression of CP T1A
predicts adverse outcomes: A potential therapeutic target for acute myeloid leukemia.
EBioMedicine 14, 5564. doi:10.1016/j.ebiom.2016.11.025
Singh, B. K., Sinha, R. A., Tripathi, M., Mendoza, A., Ohba, K., Sy, J. A. C., et al.
(2018). Thyroid hormone receptor and ERRαcoordinately regulate mitochondrial
ssion, mitophagy, biogenesis, and function. Sci. Signal 11, eaam5855. doi:10.1126/
scisignal.aam5855
Stoler, J. M., Sabry, M. A., Hanley, C., Hoppel, C. L., and Shih, V. E. (2004). Successful
long-term treatment of hepatic carnitine palmitoyltransferase I deciency and a novel
mutation. J. Inherit. Metab. Dis. 27, 679684. doi:10.1023/b:boli.0000042979.42120.55
Sun, W., Nie, T., Li, K., Wu, W., Long, Q., Feng, T., et al. (2021). Erratum. Hepatic
CPT1A facilitates liver-adipose cross talk via induction of FGF21 in mice. Diabetes
2022;71:31-42. Diabetes 71, 1827. doi:10.2337/db22-er08a
Sung, H., Ferlay, J., Siegel, R. L., Laversanne, M., Soerjomataram, I., Jemal, A., et al.
(2021). Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality
worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209249. doi:10.3322/
caac.21660
Tamura,K.,Stecher,G.,andKumar,S.(2021).MEGA11: Molecular evolutionary genetics
analysis version 11. Mol. Biol. Evol. 38, 30223027. doi:10.1093/molbev/msab120
Tan, Z., Xiao, L., Tang, M., Bai, F., Li, J., Li, L., et al. (2018). Targeting CPT1A-
mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy.
Theranostics 8, 23292347. doi:10.7150/thno.21451
Tan, Z., Zou, Y., Zhu, M., Luo, Z., Wu, T., Zheng, C., et al. (2021). Carnitine palmitoyl
transferase 1A is a novel diagnostic and predictive biomarker for breast cancer. BMC
Cancer 21, 409. doi:10.1186/s12885-021-08134-7
Tang, M., Dong, X., Xiao, L., Tan, Z., Luo, X., Yang, L., et al. (2022). CPT1A-mediated
fatty acid oxidation promotes cell proliferation via nucleoside metabolism in
nasopharyngeal carcinoma. Cell Death Dis. 13, 331. doi:10.1038/s41419-022-04730-y
Tomita, K., Tamiya, G., Ando, S., Kitamura, N., Koizumi, H., Kato, S., et al. (2005).
AICAR, an AMPK activator, has protective effects on alcohol-induced fatty liver in rats.
Alcohol Clin. Exp. Res. 29, 240S245S. doi:10.1097/01.alc.0000191126.11479.69
Torre, L. A., Bray, F., Siegel, R. L., Ferlay, J., Lortet-Tieulent, J., and Jemal, A. (2015).
Global cancer statistics, 2012. CA Cancer J. Clin. 65, 87108. doi:10.3322/caac.21262
Valentino, A., Calarco, A., Di Salle, A., Finicelli, M., Crispi, S., Calogero, R. A., et al.
(2017). Deregulation of MicroRNAs mediated control of carnitine cycle in prostate
cancer: Molecular basis and pathophysiological consequences. Oncogene 36, 60306040.
doi:10.1038/onc.2017.216
Virmani, A., Pinto, L., Bauermann, O., Zerelli, S., Diedenhofen, A., Binienda, Z. K.,
et al. (2015). The carnitine palmitoyl transferase (CPT) system and possible relevance
for neuropsychiatric and neurological conditions. Mol. Neurobiol. 52, 826836. doi:10.
1007/s12035-015-9238-7
Waks, A. G., and Winer, E. P. (2019). Breast cancer treatment: A review. JAMA 321,
288300. doi:10.1001/jama.2018.19323
Wang, C., Zhang, C., Li, X., Shen, J., Xu, Y., Shi, H., et al. (2019). CPT1A-mediated
succinylation of S100A10 increases human gastric cancer invasion. J. Cell Mol. Med . 23,
293305. doi:10.1111/jcmm.13920
Wang, D., and Lippard, S. J. (2005). Cellular processing of platinum anticancer drugs.
Nat. Rev. Drug Discov. 4, 307320. doi:10.1038/nrd1691
Wang, J., Xiang, H., Lu, Y., Wu, T., and Ji, G. (2021). The role and therapeutic
implication of CPTs in fatty acid oxidation and cancers progression. Am. J. Cancer Res.
11, 24772494.
Wang, L., Li, C., Song, Y., and Yan, Z. (2020). Inhibition of carnitine palmitoyl
transferase 1A-induced fatty acid oxidation suppresses cell progression in gastric cancer.
Arch. Biochem. Biophys. 696, 108664. doi:10.1016/j.abb.2020.108664
Wang, Y., Guo, Y. R., Liu, K., Yin, Z., Liu, R., Xia, Y., et al. (2017). KAT2A coupled
with the alpha-KGDH complex acts as a histone H3 succinyltransferase. Nature 552,
273277. doi:10.1038/nature25003
Wang, Y. N., Zeng, Z. L., Lu, J., Wang, Y., Liu, Z. X., He, M. M., et al. (2018). CPT1A-
mediated fatty acid oxidation promotes colorectal cancer cell metastasis by inhibiting
anoikis. Oncogene 37, 60256040. doi:10.1038/s41388-018-0384-z
Weber, M., Mera, P., Casas, J., Salvador, J., Rodriguez, A., Alonso, S., et al. (2020).
Liver CPT1A gene therapy reduces diet-induced hepatic steatosis in mice and highlights
potential lipid biomarkers for human NAFLD. FASEB J. 34, 1181611837. doi:10.1096/
fj.202000678R
Wen, Y. A., Xing, X., Harris, J. W., Zaytseva, Y. Y., Mitov, M. I., Napier, D. L., et al.
(2017). Adipocytes activate mitochondrial fatty acid oxidation and autophagy to
promote tumor growth in colon cancer. Cell Death Dis. 8, e2593. doi:10.1038/cddis.
2017.21
Xie, T., So, W. Y., Li, X. Y., and Leung, P. S. (2019). Fibroblast growth factor
21 protects against lipotoxicity-induced pancreatic beta-cell dysfunction via regulation
of AMPK signaling and lipid metabolism. Clin. Sci. (Lond) 133, 20292044. doi:10.1042/
CS20190093
Xiong, Y., Liu, Z., Zhao, X., Ruan, S., Zhang, X., Wang, S., et al. (2018). CPT1A
regulates breast cancer-associated lymphangiogenesis via VEGF signaling. Biomed.
Pharmacother. 106, 17. doi:10.1016/j.biopha.2018.05.112
Yao, C. H., Liu, G. Y., Wang, R., Moon, S. H., Gross, R. W., and Patti, G. J. (2018).
Identifying off-target effects of etomoxir reveals that carnitine palmitoyltransferase I is
essential for cancer cell proliferation independent of beta-oxidation. PLoS Biol. 16,
e2003782. doi:10.1371/journal.pbio.2003782
Yao, Q., Li, S., Cheng, X., Zou, Y., Shen, Y., and Zhang, S. (2020). Yin Zhi Huang, a
traditional Chinese herbal formula, ameliorates diet-induced obesity and hepatic
steatosis by activating the AMPK/SREBP-1 and the AMPK/ACC/CPT1A pathways.
Ann. Transl. Med. 8, 231. doi:10.21037/atm.2020.01.31
Yoshida, G. J. (2015). Metabolic reprogramming: The emerging concept and
associated therapeutic strategies. J. Exp. Clin. Cancer Res. 34, 111. doi:10.1186/
s13046-015-0221-y
Zeng, F., Yao, M., Wang, Y., Zheng, W., Liu, S., Hou, Z., et al. (2022). Fatty acid beta-
oxidation promotes breast cancer stemness and metastasis via the miRNA-328-3p-
CPT1A pathway. Cancer Gene Ther. 29, 383395. doi:10.1038/s41417-021-00348-y
Zhang, Y., Ma, K., Song, S., Elam, M. B., Cook, G. A., and Park, E. A. (2004).
Peroxisomal proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1 alpha)
enhances the thyroid hormone induction of carnitine palmitoyltransferase I (CPT-I
alpha). J. Biol. Chem. 279, 5396353971. doi:10.1074/jbc.M406028200
Zhang, Z., Tan, M., Xie, Z., Dai, L., Chen, Y., and Zhao, Y. (2011). Identication of
lysine succinylation as a new post-translational modication. Nat. Chem. Biol. 7, 5863.
doi:10.1038/nchembio.495
Frontiers in Pharmacology frontiersin.org14
Liang 10.3389/fphar.2023.1160440
... This study's exploration of their possible anti-metabolic syndrome effect is timely and important ( Figure 6). potential target for regulating fatty acid metabolism and understanding obesity and energy balance [44]. GLP1 (glucagon-like peptide-1) affects insulin secretion, appetite, and stomach emptying, suggesting that controlling GLP1 could significantly impact glucose control and weight management [45,46]. ...
... Its inhibition helps treat dyslipidemia by decreasing the production of cholesterol in the body [43]. CPT-1, which plays a role in the mitochondrial breakdown of long-chain fatty acids, is a potential target for regulating fatty acid metabolism and understanding obesity and energy balance [44]. GLP1 (glucagon-like peptide-1) affects insulin secretion, appetite, and stomach emptying, suggesting that controlling GLP1 could significantly impact glucose control and weight management [45,46]. ...
... Quinones enhance the therapeutic potential of E. bulbosa against metabolic syndrome due to their strong antioxidant and anti-inflammatory properties [49]. The ability of FOE to decrease the expression of important proteins such as CPT-1 and boost GLP1 activity explains its antidiabetic and lipid-lowering benefits [44,48,49]. ...
Article
Full-text available
Metabolic syndrome is a global health problem. The use of functional foods as dietary components has been increasing. One food of interest is forest onion extract (FOE). This study aimed to investigate the effect of FOE on lipid and glucose metabolism in silico and in vitro using the 3T3-L1 mouse cell line. This was a comprehensive study that used a multi-modal computational network pharmacology analysis and molecular docking in silico and 3T3-L1 mouse cells in vitro. The phytochemical components of FOE were analyzed using untargeted ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS). Next, an in silico analysis was performed to determine FOE’s bioactive compounds, and a toxicity analysis, protein target identification, network pharmacology, and molecular docking were carried out. FOE’s effect on pancreatic lipase, α-glucosidase, and α-amylase inhibition was determined. Finally, we determined its effect on lipid accumulation and MAPK8, PPARG, HMGCR, CPT-1, and GLP1 expression in the preadipocyte 3T3-L1 mouse cell line. We showed that the potential metabolites targeted glucose and lipid metabolism in silico and that FOE inhibited pancreatic lipase levels, α-glucosidase, and α-amylase in vitro. Furthermore, FOE significantly (p < 0.05) inhibits targeted protein expressions of MAPK8, PPARG, HMGCR, CPT-1, and GLP-1 in vitro in 3T3-L1 mouse cells in a dose-dependent manner. FOE contains several metabolites that reduce pancreatic lipase levels, α-glucosidase, α-amylase, and targeted proteins associated with lipid and glucose metabolism in vitro.
... Having established that TSPO deficiency modulates FAO in mouse primary astrocytes, we began to explore the potential underlying mechanism. CPT1a, the rate-limiting enzyme of FAO (Houten et al., 2016;Schlaepfer & Joshi, 2020), resides in the outer mitochondrial membrane (Liang, 2023). Previous work has established that CPT1a forms part of a complex with another outer mitochondrial membrane protein, the voltage-dependent anion channel (VDAC) (Lee et al., 2011;Liang, 2023). ...
... CPT1a, the rate-limiting enzyme of FAO (Houten et al., 2016;Schlaepfer & Joshi, 2020), resides in the outer mitochondrial membrane (Liang, 2023). Previous work has established that CPT1a forms part of a complex with another outer mitochondrial membrane protein, the voltage-dependent anion channel (VDAC) (Lee et al., 2011;Liang, 2023). VDAC is known to form complexes with TSPO (Gatliff et al., 2014), but the existence of a protein complex containing TSPO and CPT1a has not yet been experimentally confirmed. ...
Article
Full-text available
The mitochondrial translocator protein 18 kDa (TSPO) has been linked to functions from steroidogenesis to regulation of cellular metabolism and is an attractive therapeutic target for chronic CNS inflammation. Studies in Leydig cells and microglia indicate that TSPO function may vary between cells depending on their specialized roles. Astrocytes are critical for providing trophic and metabolic support in the brain. Recent work has highlighted that TSPO expression increases in astrocytes under inflamed conditions and may drive astrocyte reactivity. Relatively little is known about the role TSPO plays in regulating astrocyte metabolism and whether this protein is involved in immunometabolic processes in these cells. Using TSPO‐deficient (TSPO−/−) mouse primary astrocytes in vitro (MPAs) and a human astrocytoma cell line (U373 cells), we performed extracellular metabolic flux analyses. We found that TSPO deficiency reduced basal cellular respiration and attenuated the bioenergetic response to glucopenia. Fatty acid oxidation was increased, and lactate production was reduced in TSPO−/− MPAs and U373 cells. Co‐immunoprecipitation studies revealed that TSPO forms a complex with carnitine palmitoyltransferase 1a in U373 and MPAs, presenting a mechanism wherein TSPO may regulate FAO in these cells. Compared to TSPO+/+ cells, in TSPO−/− MPAs we observed attenuated tumor necrosis factor release following 3 h lipopolysaccharide (LPS) stimulation, which was enhanced at 24 h post‐LPS stimulation. Together these data suggest that while TSPO acts as a regulator of metabolic flexibility, TSPO deficiency does not appear to modulate the metabolic response of MPAs to inflammation, at least in response to the model used in this study. image
... PPARα and PGC1α could enhance CPT1A expression at transcriptional level. miR-33a/b, miR-124, miR-328-3p, or miR-370 inhibits the translation of CPT1A to suppress fatty acid β-oxidation [37]. In addition, the uptake of fatty acids is also important for fatty acid degradation. ...
Article
Full-text available
Congenital heart disease (CHD) is the most serious form of heart disease, and chronic hypoxia is the basic physiological process underlying CHD. Some patients with CHD do not undergo surgery, and thus, they remain susceptible to chronic hypoxia, suggesting that some protective mechanism might exist in CHD patients. However, the mechanism underlying myocardial adaptation to chronic hypoxia remains unclear. Proteomics was used to identify the differentially expressed proteins in cardiomyocytes cultured under hypoxia for different durations. Western blotting assays were used to verify protein expression. A Real-Time Cell Analyzer (RTCA) was used to analyze cell growth. In this study, 3881 proteins were identified by proteomics. Subsequent bioinformatics analysis revealed that proteins were enriched in regulating oxidoreductase activity. Functional similarity cluster analyses showed that chronic hypoxia resulted in proteins enrichment in the mitochondrial metabolic pathway. Further KEGG analyses found that the proteins involved in fatty acid metabolism, the TCA cycle and oxidative phosphorylation were markedly upregulated. Moreover, knockdown of CPT1A or ECI1, which is critical for fatty acid degradation, suppressed the growth of cardiomyocytes under chronic hypoxia. The results of our study revealed that chronic hypoxia activates fatty acid metabolism to maintain the growth of cardiomyocytes.
... Inhibition of CPT-I can distort fatty acid transport across the mitochondrial membrane, leading to severe side effects such as FAO inhibition and hepatic steatosis [13,22,48]. Valproate restricts FAO by interacting with the acyl-CoA formation. ...
Article
Full-text available
Drug induced fatty liver disease (DIFLD) is a form of drug-induced liver injury (DILI), which can also be included in the more general metabolic dysfunction-associated steatotic liver disease (MASLD), which specifically refers to the accumulation of fat in the liver unrelated to alcohol intake. A bi-directional relationship between DILI and MASLD is likely to exist: while certain drugs can cause MASLD by acting as pro-steatogenic factors, MASLD may make hepatocytes more vulnerable to drugs. Having a pre-existing MASLD significantly heightens the likelihood of experiencing DILI from certain medications. Thus, the prevalence of steatosis within DILI may be biased by pre-existing MASLD, and it can be concluded that the genuine true incidence of DIFLD in the general population remains unknown. In certain individuals, drug-induced steatosis is often accompanied by concomitant injury mechanisms such as oxidative stress, cell death, and inflammation, which leads to the development of drug-induced steatohepatitis (DISH). DISH is much more severe from the clinical point of view, has worse prognosis and outcome, and resembles MASH (metabolic-associated steatohepatitis), as it is associated with inflammation and sometimes with fibrosis. A literature review of clinical case reports allowed us to examine and evaluate the clinical features of DIFLD and their association with specific drugs, enabling us to propose a classification of DIFLD drugs based on clinical outcomes and pathological severity: Group 1, drugs with low intrinsic toxicity (e.g., ibuprofen, naproxen, acetaminophen, irinotecan, methotrexate, and tamoxifen), but expected to promote/aggravate steatosis in patients with pre-existing MASLD; Group 2, drugs associated with steatosis and only occasionally with steatohepatitis (e.g., amiodarone, valproic acid, and tetracycline); and Group 3, drugs with a great tendency to transit to steatohepatitis and further to fibrosis. Different mechanisms may be in play when identifying drug mode of action: (1) inhibition of mitochondrial fatty acid β-oxidation; (2) inhibition of fatty acid transport across mitochondrial membranes; (3) increased de novo lipid synthesis; (4) reduction in lipid export by the inhibition of microsomal triglyceride transfer protein; (5) induction of mitochondrial permeability transition pore opening; (6) dissipation of the mitochondrial transmembrane potential; (7) impairment of the mitochondrial respiratory chain/oxidative phosphorylation; (8) mitochondrial DNA damage, degradation and depletion; and (9) nuclear receptors (NRs)/transcriptomic alterations. Currently, the majority of, if not all, adverse outcome pathways (AOPs) for steatosis in AOP-Wiki highlight the interaction with NRs or transcription factors as the key molecular initiating event (MIE). This perspective suggests that chemical-induced steatosis typically results from the interplay between a chemical and a NR or transcription factors, implying that this interaction represents the primary and pivotal MIE. However, upon conducting this exhaustive literature review, it became evident that the current AOPs tend to overly emphasize this interaction as the sole MIE. Some studies indeed support the involvement of NRs in steatosis, but others demonstrate that such NR interactions alone do not necessarily lead to steatosis. This view, ignoring other mitochondrial-related injury mechanisms, falls short in encapsulating the intricate biological mechanisms involved in chemically induced liver steatosis, necessitating their consideration as part of the AOP’s map road as well.
... In addition, peroxisome proliferator-activated receptor alpha (PPARα) is a nuclear receptor that plays a crucial role in the regulation of lipid metabolism, inflammation, and oxidative stress. Concurrently, carnitine palmitoyl transferase 1A (CPT1A) as a downstream target of PPARα, is a central regulatory factor for β-oxidation of free fatty acids in mitochondria [14]. Furthermore, studies evidence that dysregulation of PPARα in NAFLD, suggesting a potential involvement in disease pathogenesis [15,16]. ...
Article
Full-text available
Background Non-alcoholic fatty liver disease (NAFLD) is a multifaceted metabolic disorder, whose global prevalence is rapidly increasing. Acetyl CoA carboxylases 1 (ACACA) is the key enzyme that controls the rate of fatty acid synthesis. Hence, it is crucial to investigate the function of ACACA in regulating lipid metabolism during the progress of NAFLD. Methods Firstly, a fatty liver mouse model was established by high-fat diet at 2nd, 12th, and 20th week, respectively. Then, transcriptome analysis was performed on liver samples to investigate the underlying mechanisms and identify the target gene of the occurrence and development of NAFLD. Afterwards, lipid accumulation cell model was induced by palmitic acid and oleic acid (PA ∶ OA molar ratio = 1∶2). Next, we silenced the target gene ACACA using small interfering RNAs (siRNAs) or the CMS-121 inhibitor. Subsequently, experiments were performed comprehensively the effects of inhibiting ACACA on mitochondrial function and lipid metabolism, as well as on AMPK- PPARα- CPT1A pathway. Results This data indicated that the pathways significantly affected by high-fat diet include lipid metabolism and mitochondrial function. Then, we focus on the target gene ACACA. In addition, the in vitro results suggested that inhibiting of ACACA in vitro reduces intracellular lipid accumulation, specifically the content of TG and TC. Furthermore, ACACA ameliorated mitochondrial dysfunction and alleviate oxidative stress, including MMP complete, ATP and ROS production, as well as the expression of mitochondria respiratory chain complex (MRC) and AMPK proteins. Meanwhile, ACACA inhibition enhances lipid metabolism through activation of PPARα/CPT1A, leading to a decrease in intracellular lipid accumulation. Conclusion Targeting ACACA can reduce lipid accumulation by mediating the AMPK- PPARα- CPT1A pathway, which regulates lipid metabolism and alleviates mitochondrial dysfunction.
Article
Full-text available
Succinylation modification involves in the progression of human cancers. The present study aimed to investigate the role of CPT1A, which is a succinyltransferase in the progression of prostate cancer (PCa). CCK-8 was used to detect the cell viability. Seahorse was performed to evaluate the cell glycolysis. Luciferase assay was used to detect the transcriptional regulation. ChIP was performed to assess the binding between transcriptional factors with the promoters. Co-IP was used to assess the binding between proteins. We found that CPT1A was highly expressed in PCa tissues and cell lines. Silencing of CPT1A inhibited the viability and glycolysis of PCa cells. Mechanistically, CPT1A promoted the succinylation of SP5, which strengthened the binding between SP5 and the promoter of PDPK1. SP5 activated PDPK1 transcription and PDPK1 activated the AKT/mTOR signal pathway. These findings might provide novel targets for the diagnosis or therapy of prostate cancer.
Article
Full-text available
PurposeLimited treatment options are currently available for glioblastoma (GBM), an extremely lethal type of brain cancer. For a variety of tumor types, bioenergetic deprivation through inhibition of cancer-specific metabolic pathways has proven to be an effective therapeutic strategy. Here, we evaluated the therapeutic effects and underlying mechanisms of dual inhibition of carnitine palmitoyltransferase 1A (CPT1A) and glucose-6-phosphate dehydrogenase (G6PD) critical for fatty acid oxidation (FAO) and the pentose phosphate pathway (PPP), respectively, against GBM tumorspheres (TSs).Methods Therapeutic efficacy against GBM TSs was determined by assessing cell viability, neurosphere formation, and 3D invasion. Liquid chromatography-mass spectrometry (LC–MS) and RNA sequencing were employed for metabolite and gene expression profiling, respectively. Anticancer efficacy in vivo was examined using an orthotopic xenograft model.ResultsCPT1A and G6PD were highly expressed in GBM tumor tissues. Notably, siRNA-mediated knockdown of both genes led to reduced viability, ATP levels, and expression of genes associated with stemness and invasiveness. Similar results were obtained upon combined treatment with etomoxir and dehydroepiandrosterone (DHEA). Transcriptome analyses further confirmed these results. Data from LC–MS analysis showed that this treatment regimen induced a considerable reduction in the levels of metabolites associated with the TCA cycle and PPP. Additionally, the combination of etomoxir and DHEA inhibited tumor growth and extended survival in orthotopic xenograft model mice.Conclusion Our collective findings support the utility of dual suppression of CPT1A and G6PD with selective inhibitors, etomoxir and DHEA, as an efficacious therapeutic approach for GBM.
Article
Full-text available
As the first rate-limiting enzyme in fatty acid oxidation (FAO), CPT1 plays a significant role in metabolic adaptation in cancer pathogenesis. FAO provides an alternative energy supply for cancer cells and is required for cancer cell survival. Given the high proliferation rate of cancer cells, nucleotide synthesis gains prominence in rapidly proliferating cells. In the present study, we found that CPT1A is a determining factor for the abnormal activation of FAO in nasopharyngeal carcinoma (NPC) cells. CPT1A is highly expressed in NPC cells and biopsies. CPT1A dramatically affects the malignant phenotypes in NPC, including proliferation, anchorage-independent growth, and tumor formation ability in nude mice. Moreover, an increased level of CPT1A promotes core metabolic pathways to generate ATP, inducing equivalents and the main precursors for nucleotide biosynthesis. Knockdown of CPT1A markedly lowers the fraction of ¹³ C-palmitate-derived carbons into pyrimidine. Periodic activation of CPT1A increases the content of nucleoside metabolic intermediates promoting cell cycle progression in NPC cells. Targeting CPT1A-mediated FAO hinders the cell cycle G1/S transition. Our work verified that CPT1A links FAO to cell cycle progression in NPC cellular proliferation, which supplements additional experimental evidence for developing a therapeutic mechanism based on manipulating lipid metabolism.
Article
Full-text available
Glioblastoma multiforme (GBM) remains the top challenge to radiotherapy with only 25% one-year survival after diagnosis. Here, we reveal that co-enhancement of mitochondrial fatty acid oxidation (FAO) enzymes (CPT1A, CPT2 and ACAD9) and immune checkpoint CD47 is dominant in recurrent GBM patients with poor prognosis. A glycolysis-to-FAO metabolic rewiring is associated with CD47 anti-phagocytosis in radioresistant GBM cells and regrown GBM after radiation in syngeneic mice. Inhibition of FAO by CPT1 inhibitor etomoxir or CRISPR-generated CPT1A−/−, CPT2−/−, ACAD9−/− cells demonstrate that FAO-derived acetyl-CoA upregulates CD47 transcription via NF-κB/RelA acetylation. Blocking FAO impairs tumor growth and reduces CD47 anti-phagocytosis. Etomoxir combined with anti-CD47 antibody synergizes radiation control of regrown tumors with boosted macrophage phagocytosis. These results demonstrate that enhanced fat acid metabolism promotes aggressive growth of GBM with CD47-mediated immune evasion. The FAO-CD47 axis may be targeted to improve GBM control by eliminating the radioresistant phagocytosis-proofing tumor cells in GBM radioimmunotherapy. Acquired radioresistance is a challenge for the cure of glioblastoma. Here, the authors show that radioresistant glioblastoma boosts mitochondrial fatty acid oxidation that fuels cell proliferation and induces immunosuppression via CD47 mediated anti-phagocytosis. Inhibition of FAO by etomoxir combined with anti-CD47 antibodies sensitizes glioblastoma to radiotherapy.
Article
Full-text available
Nonalcoholic fatty liver disease (NAFLD) is one of the most prevalent forms of chronic liver diseases and is causally linked to hepatic insulin resistance and reduced fatty acid oxidation. Therapeutic treatments targeting both hepatic insulin resistance and lipid oxidative metabolism are considered as feasible strategies to alleviate this disease. Emerging evidence suggests Estrogen-Related Receptor alpha (ERRα), the first orphan nuclear receptor identified, as a master regulator in energy homeostasis by controlling glucose and lipid metabolism. Small molecules improving the functions of ERRα may provide a new option for management of NAFLD. In the present study, by using liver-specific Errα knockout mouse (Errα-LKO), we showed that liver-specific deletion of ERRα exacerbated diet-evoked fatty liver, hepatic and systemic insulin resistance in mice. A potent and selective ERRα agonist JND003 (7) was also discovered. In vitro and in vivo investigation demonstrated that the compound enhanced the transactivation of ERRα downstream target genes, which was accompanied by improved insulin sensitivity and fatty liver symptoms. Furthermore, the therapeutic effects were completely abolished in Errα-LKO mice, indicative of its on-target efficacy. Our study thus suggests that hepatic ERRα is a viable target for NAFLD and that ERRα agonist may serve as an intriguing pharmacological option for management of metabolic diseases.
Article
Full-text available
Cancer cells must maintain metabolic homeostasis under a wide range of conditions and meet their own energy needs in order to survive and reproduce. In addition to glycolysis, cancer cells can also perform various metabolic strategies, such as fatty acid oxidation (FAO). It has been found that the proliferation, survival, drug resistance and metastasis of cancer cells depend on FAO. The carnitine palmitoyltransferase (CPT), including CPT1 and CPT2, located on the mitochondrial membrane, are important mediators of FAO. In recent years, many researchers have found that CPT has a close relationship with the metabolic development of tumor cells, not only provides energy for cancer cells development and metastasis by promoting FAO but also affects the occurrence and invasion through other signal pathways or cytokines or microRNA. This review summarized the role of CPTs in several kinds of tumors and the developed targeted inhibitors of CPTs, as well as the potential gene therapy and immunotherapy of CPTs, hoping to better explore the mechanism and role of CPTs in the future and providing useful ideas for clinical treatment.
Article
Full-text available
MicroRNAs (miRNA) have been shown to be associated with tumor diagnosis, prognosis, and therapeutic response. MiR-328-3p plays a significant role in breast cancer growth; however, its actual function and how it modulates specific biological functions is poorly understood. Here, miR-328-3p was significantly downregulated in breast cancer, especially in patients with metastasis. Mitochondrial carnitine palmitoyl transferase 1a (CPT1A) is a downstream target gene in the miR-328-3p-regulated pathway. Furthermore, the miR-328-3p/CPT1A/fatty acid β-oxidation/stemness axis was shown responsible for breast cancer metastasis. Collectively, this study revealed that miR-328-3p is a potential therapeutic target for the treatment of breast cancer patients with metastasis, and also a model for the miRNA-fatty acid β-oxidation-stemness axis, which may assist inunderstanding the cancer stem cell signaling functions of miRNA.
Article
In Fig. 3B of article cited above, for images of iWAT and eWAT, the labels “WT” and “LKO” were inadvertently transposed. LKO (Cpt1a knockout) should have been the label for the first set of images of adipose tissue, and WT (wild type) should have been the label for the second set of images. The authors apologize for the error. The online version of the article (https://doi.org/10.2337/db21-0363) has been updated to correct the error.
Article
Glioma stem cells (GSCs) are self-renewing tumor cells with multi-lineage differentiation potential and the capacity of construct glioblastoma (GBM) heterogenicity. Mitochondrial morphology is associated with the metabolic plasticity of GBM cells. Previous studies have revealed distinct mitochondrial morphologies and metabolic phenotypes between GSCs and non-stem tumor cells (NSTCs), whereas the molecules regulating mitochondrial dynamics in GBM cells are largely unknown. Herein, we report that carnitine palmitoyltransferase 1A (CPT1A) is preferentially expressed in NSTCs, and governs mitochondrial dynamics and GSC differentiation. Expressions of CPT1A and GSC marker CD133 were mutually exclusive in human GBMs. Overexpression of CPT1A inhibited GSC self-renewal but promoted mitochondrial fusion. In contrast, disruption of CPT1A in NSTCs promoted mitochondrial fission and reprogrammed NSTCs toward GSC feature. Mechanistically, CPT1A overexpression increased the phosphorylation of dynamin-related protein 1 at Ser-637 to promote mitochondrial fusion. In vivo, CPT1A overexpression decreased the percentage of GSCs, impaired GSC-derived xenograft growth and prolonged tumor-bearing mice survival. Our work identified CPT1A as a critical regulator of mitochondrial dynamics and GSC differentiation, indicating that CPT1A could be developed as a molecular target for GBM cell-differentiation strategy. The authors demonstrate that carnitine palmitoyltransferase 1A (CPT1A) expression is reduced in glioma stem cells (GSCs) in comparison with non-stem tumor cells. CPT1A overexpression promotes mitochondrial fusion and GSC differentiation by increasing the phosphorylation of dynamin-related protein 1 (Drp1) at Ser-637, thus impairing GSC-derived xenograft growth and prolonging survival in tumor-bearing mice. These results suggest that CPT1A could be a molecular target for GSC differentiation therapy.
Article
Fatty acid metabolism is known to support tumorigenesis and disease progression as well as treatment resistance through enhanced lipid synthesis, storage and catabolism. More recently, the role of membrane fatty acid composition, for example, ratios of saturated, monounsaturated and polyunsaturated fatty acids, in promoting cell survival while limiting lipotoxicity and ferroptosis has been increasingly appreciated. Alongside these insights, it has become clear that tumour cells exhibit plasticity with respect to fatty acid metabolism, responding to extratumoural and systemic metabolic signals, such as obesity and cancer therapeutics, to promote the development of aggressive, treatment-resistant disease. Here, we describe cellular fatty acid metabolic changes that are connected to therapy resistance and contextualize obesity-associated changes in host fatty acid metabolism that likely influence the local tumour microenvironment to further modify cancer cell behaviour while simultaneously creating potential new vulnerabilities.