You are on page 1of 15

Cell Metabolism

Review

Obesity and Cancer: The Oil that Feeds the Flame


Joan Font-Burgada,1 Beicheng Sun,2,* and Michael Karin1,*
1Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, Moores Cancer Center, UCSD

School of Medicine, La Jolla, CA 92093-0723, USA


2Liver Transplantation Center of the First Affiliated Hospital and Cancer Center, Nanjing Medical University, Nanjing, Jiangsu Province, P.R.

China
*Correspondence: sunbc@njmu.edu.cn (B.S.), karinoffice@ucsd.edu (M.K.)
http://dx.doi.org/10.1016/j.cmet.2015.12.015

Although discussion of the obesity epidemic had become a cocktail party cliché, its impact on public health
cannot be dismissed. In the past decade, cancer had joined the list of chronic debilitating diseases whose
risk is substantially increased by hypernutrition. Here we discuss recent advances in understanding how
obesity increases cancer risk and propose a unifying hypothesis according to which the major tumor-pro-
moting mechanism triggered by hypernutrition is the indolent inflammation that takes place at particular
organ sites, including liver, pancreas, and gastrointestinal tract. The mechanisms by which excessive fat
deposition feeds this tumor-promoting inflammatory flame are diverse and tissue specific.

Obesity and Cancer tion, 2004). In 2011–2012, it was estimated that over one-third of
Cancer is a complex disease with genetic, environmental, U.S. adults were obese, but certain ethnic groups are at much
clinical, and lifestyle factors, all of which contribute to tumor higher risk of obesity-related morbidities than the average.
initiation and malignant progression. Tobacco smoke and sun Recently, obesity trends have markedly increased in developing
exposure are two of the most extensively studied lifestyle- nations, particularly in children, although traditionally it was
related factors that greatly and directly increase cancer risk, considered as a problem unique to adults in affluent societies
but the carcinogenic impact of obesity is nearly as profound (Ogden et al., 2014). Notably, the incidence of obesity in rapidly
and has become more prevalent, as obesity rates in major parts developing countries (Middle East, Oceania, Australasia, and
of the world are beginning to eclipse smoking rates. Epidemio- China) has risen 3-fold or more since 1980 (Ellulu et al., 2014),
logic evidence linking obesity and hypernutrition to elevated can- and currently the rate of obesity in Mexico is higher than in
cer incidence and greater risk of cancer-related death has accu- most of the US.
mulated in the past two decades, leading to the realization that In addition to classical metabolic diseases, in particular type 2
14% and 20% of all cancer deaths in men and women, respec- diabetes (T2DM) and cardiovascular disease (Ng et al., 2014),
tively, are due to overweight and obesity (Calle et al., 2003). obesity increases cancer risk. A significant association between
Despite the huge impact, the molecular mechanisms explaining obesity and cancer at various sites was detected more than a
this association remain poorly understood and their investigation decade ago and it was estimated that the overall risk of death
was only recently initiated. from cancer is 1.5- to 1.6-fold higher in men and women, respec-
In nature, food availability is not uniform and a constant caloric tively, with a BMI that exceeds 40 kg/m2 higher (Calle et al.,
input is rare, other than under laboratory conditions or in highly 2003). At certain sites, such as pancreas and liver in obese
affluent societies. All eukaryotes, including plants, store energy men, the risk of developing deadly cancer is elevated by up to
in the form of lipids and complex carbohydrates. Energy storage 2.6- and 4.5-fold, respectively, whereas obese women exhibit
is key to physiological homeostasis and essential for maintaining a 4.8- and 5.3-fold increase in risk of death due to kidney and in-
health and reproductive potential, thereby contributing to the testinal cancer, respectively (Calle and Kaaks, 2004; Haslam and
major driving force of evolution—survival of the species. How- James, 2005). Obesity triggers several systemic and metabolic
ever, in recent decades, humans have constantly increased alterations that can influence carcinogenesis and tumorigenesis
their caloric intake with a concomitant drop in energy expendi- that will be summarized and discussed in this Review (Figure 1).
ture due to a more sedentary lifestyle. The result of this net Clearly, obesity does not influence cancer risk to the same
gain in caloric input and access to a constant supply of en- extent in different organs, and the tumor-promoting mechanisms
ergy-rich foods is a metabolic imbalance accompanied by hy- evoked by obesity are not only tissue specific but are also gender
pertrophy and hyperplasia of adipose tissue and consequent specific. We will mainly focus our discussion on gastrointestinal,
obesity, associated with fat deposition outside of conventional pancreatic, and liver cancers.
lipid storage depots. The pathophysiological impact of obesity
far exceeds its cosmetic effect on body shape. Systemic Consequences of Obesity that Promote
People are defined as obese when their body mass index Tumorigenesis
(BMI) exceeds 30 kg/m2 and overweight when it is between 25 Systemic Inflammation
and 30 kg/m2 (Rocchini, 2002). BMI, however, is not an accurate In addition to being hyperplastic and hypertrophic, the obese ad-
measure of central obesity across all populations, as Asian ipose tissue exhibits major changes in production of hormones,
Americans and East Asians, for instance, experience central adipokines, and cytokines, as well as altered immune infiltration
obesity at a lower BMI than Caucasians (WHO Expert Consulta- (Hotamisligil et al., 1993; Weisberg et al., 2003; Xu et al., 2003)

48 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.


Cell Metabolism

Review

Figure 1. Systemic Mechanisms of Obesity-Driven Tumor Promotion


Obesity and hypernutrition affect different tissues simultaneously resulting in a systemic increase in NEFA, insulin, glucose, leptin and inflammatory cytokines,
and reduced levels of adiponectin. These imbalances can directly promote cancer cell survival, proliferation, and malignant progression. In the colon, obesity can
promote a shift in the composition of the microbiota that is linked to higher epithelial permeability to microbial components such as LPS, which in turn enhances
local inflammation. In adipose tissue, obesity is linked to adipocyte hyperplasia and hypertrophy followed by infiltration of inflammatory macrophages. The
inflammatory milieu thus generated reprograms the tissue to release NEFA, produce more leptin, and less adiponectin. The liver, exposed to microbial products
from the colon via the portal circulation, also mounts a low-grade inflammatory response. Higher amounts of NEFA from adipose tissue lead to an increased lipid
storage and elevated lipogenesis. The consequence of local inflammation and lipid accumulation is liver insulin resistance and hepatocyte stress and death.
Finally, the pancreas reacts to hepatic insulin resistance and higher glycemia with increased insulin production, which can lead to b-cell stress.

(Figure 1). Although numerous immune cell types are present in may also enhance DNA damage, thus increasing mutational load
fat depots, macrophages are especially common and their prop- (Yan et al., 2006). TNF also induces epithelial to mesenchymal
erties are altered by obesity (Weisberg et al., 2003). Adipose tis- transition (EMT), which provides another route for stimulating
sue macrophages (ATM) in healthy fat depots are skewed toward malignancy and metastatic behavior (Kim et al., 2009). IL-6 acti-
the M2 anti-inflammatory phenotype, but during obesity, pro-in- vates STAT3, which controls expression of genes that promote
flammatory M1 macrophages become more abundant (McNelis cell proliferation, survival, angiogenesis, invasiveness, metas-
and Olefsky, 2014). Pro-inflammatory ATM produce tumor-pro- tasis, and stemness (Taniguchi and Karin, 2014). IL-1b exerts
moting cytokines, including TNF, IL-6, and IL-1b, as well as effects similar to those of TNF and all three cytokines, whose
monocyte chemo-attractants such as MCP-1, CCL-2, and MIF. expression is elevated during obesity, have well-established
Elevated serum IL-6 and TNF are typically found in obese pa- tumor-promoting activities (Grivennikov et al., 2010). Other
tients (Weisberg et al., 2003), and the signaling circuitry respon- tumor-promoting cytokines whose expression was noted to be
sible for their production is orchestrated by the IKK–NF-kB and elevated during obesity include IL-8, IL-10, IL-12, IL-17, IL-18,
JNK–AP-1 pathways (Solinas et al., 2007). The initial activation IL-22, and interferon (IFN)-g. Importantly, protective anti-inflam-
of these pathways seems to depend on damage-associated mo- matory Treg cells that are specifically located in abdominal fat
lecular patterns (DAMPs) secreted by stressed and dying adipo- depots are lost in obesity, further exacerbating the pro-inflam-
cytes and on changes in membrane fluidity caused by uptake matory state associated with elevated BMI (Feuerer et al., 2009).
and incorporation of saturated fatty acids (Holzer et al., 2011). Adipokines
In addition, ATM undergo changes in lysosome-dependent lipid Adipokines are hormones and cytokines that are produced by
metabolism that favor lipid accumulation (Xu et al., 2013). adipocytes and exert diverse and important effects on meta-
TNF signaling in cancer cells enhances survival and prolifera- bolism, angiogenesis, and cell proliferation. Adiponectin, en-
tion through NF-kB and JNK activation (Karin and Lin, 2002). TNF coded by the Ad/Poq gene, is secreted by adipocytes and binds

Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc. 49


Cell Metabolism

Review

to two receptors (AdipoR1 and 2) that differ in their tissue distri- physical activity give rise to favorable IL-6 plasma concentra-
bution but are frequently expressed by cancer cells. Most exper- tions in obese children and adolescents (Gallistl et al., 2001).
imental data point to adiponectin as an inhibitor of tumorigenesis Both IL-6 and TNF elevate the hepatic and circulating concentra-
due to its ability to activate AMPK and inhibit mTORC1 and other tions of C-reactive protein (CRP), which is a nonspecific inflam-
protumorigenic signaling molecules (Dalamaga et al., 2012). matory response marker that is frequently increased in obesity.
Congruently, adiponectin expression is inversely associated Two important tumor-promoting effects are derived from
with obesity (Antuna-Puente et al., 2008) (Figure 1). Adiponectin obesity-induced IR: hyperinsulinemia and hyperglycemia. High
enhances insulin sensitivity by activating AMPK and reducing concentrations of insulin, glucose, and NEFA are strong pro-
inflammation (Yamauchi et al., 2002). Importantly, weight loss in- moters of cell survival, growth, and proliferation and exert similar
creases the amounts of circulating adiponectin in obese individ- effects on tumor progenitors. High glucose concentrations favor
uals (Reinehr et al., 2004). Leptin is an adipokine that was mainly glycolytic cancer cell metabolism characterized by enhanced
studied for its appetite-regulating functions exerted in the hypo- glucose consumption (Warburg, 1956). Hyperglycemia also re-
thalamus. Obese individuals produce higher amounts of leptin sults in activation of Hif1a, which increases survival of cancer
due to their larger adipose tissue volume (Considine et al., cells under hypoxic conditions and further enhances expression
1996) (Figure 1), but this can result in central (hypothalamic) lep- of glycolytic enzymes (Catrina et al., 2004). Hyperinsulinemia ex-
tin resistance (El-Haschimi et al., 2000). The main pro-tumori- erts additional tumor-promoting effects due to the intrinsic
genic effect of leptin is attributed to its growth factor-like activity growth stimulatory and antiapoptotic effects of insulin. Interest-
on endothelial cells, resulting in enhanced angiogenesis (Uddin ingly, dietary sugar intake was found to enhance liver tumorigen-
et al., 2011). Like IL-6, leptin activates STAT3, which provides esis in mice independently of excess adiposity (Healy et al.,
another explanation for its pro-tumorigenic activity. Other mole- 2015).
cules considered adipokines with possible contribution to can- Dysbiosis
cer are chemelin, apellin, PAI-1, FGF-21, omentin, and lipocalin Consumption of foods with high fat and low fiber content can
(Fasshauer and Blüher, 2015). elicit particular changes in the microflora inhabiting the human
Insulin Resistance and Hyperglycemia digestive tract that lead to decreased microbiome diversity and
Obesity is associated with the metabolic syndrome, which in- dysbiosis (Figure 1). Such alterations were linked to a number
cludes insulin resistance (IR), T2DM, atherosclerosis, and high of diseases, including cancer (Schwabe and Jobin, 2013). During
blood pressure. Conversely, weight loss markedly improves in- obesity, the composition of the gut microbiota and the properties
sulin sensitivity and reduces insulin production (Uusitupa et al., of the intestinal epithelium are altered, resulting in decreased
2003). IR is a physiological condition defined by target cells barrier function (Teixeira et al., 2012; Turnbaugh et al., 2006).
that are resistant to insulin action when encountering insulin Obesity can also cause overrepresentation of bacterial species
within the normal concentration range. IR muscle cells are that produce pro-carcinogenic metabolites (Louis et al., 2014),
compromised in glucose uptake and metabolism, resulting in although the clinical significance of such metabolites remains
hyperglycemia, which exposes other cells, including malignant to be established. Dysbiosis alters metabolic usage and can
cells, to higher than normal glucose concentrations. Obesity trig- aggravate the metabolic imbalance already present in obese in-
gers IR by diverse mechanisms, but it primarily acts through dividuals (Tilg and Kaser, 2011). Dysbiosis also alters the gut
elevated non-esterified fatty acids (NEFAs), also called free fatty epithelial barrier, making it more permeable to microbial prod-
acids (FFAs), which arise from adipocytes. Of note, individuals ucts that trespass the mucosa and activate immune cells in the
subjected to an acute increase in NEFA develop IR within hours lamina propria, as well as reach the liver via the portal circulation,
(Roden et al., 1996). High saturated NEFA concentrations result all of which enhance systemic inflammation and contribute to
in inhibitory phosphorylation of IRS-1 and -2 and suppression production of pro-inflammatory cytokines, such as TNF and IL-
of insulin signaling (Aguirre et al., 2002; Solinas et al., 2006). 6, and to IR (Shi et al., 2006). Recently, it was shown that
Not all fat depots have the same influence on IR; intra-abdominal commonly used emulsifiers present in processed foods also
fat has a higher lipolytic activity compared to peripheral fat and alter the intestinal microbiota and elicit a metabolic syndrome
its close proximity to the liver can explain the stronger increase phenotype (Chassaing et al., 2015), demonstrating that dysbio-
in liver IR in obese individuals (Nielsen et al., 2004). The systemic sis can have different origins. Uncontrolled antibiotic use can
increase in inflammatory cytokines also contributes to IR. TNF also lead to dysbiosis and obesity (Cho and Blaser, 2012) and
and IL-6 increase adipocyte lipolytic activity (Yang et al., 2008; that may increase cancer risk later in life. Increased barrier
Zhang et al., 2002) and thus increase NEFA concentrations permeability was shown to be a major contributor to colorectal
concomitantly with reduced insulin receptor signaling (Figure 1). tumorigenesis (Grivennikov et al., 2012) and can also affect the
By activating JNK, TNF promotes inhibitory phosphorylation of liver and pancreas (Tremaroli and Bäckhed, 2012; Yoshimoto
IRS-1 (Hirosumi et al., 2002) and IRS-2 (Solinas et al., 2006). et al., 2013).
Conversely, TNF-deficient mice are protected from obesity- Immune Dysregulation
related IR (Uysal et al., 1997). Administration of exogenous Obesity has been linked to an impaired immune response that
TNF causes IR and its neutralization reverses IR (Cheung et al., acts as a risk factor for different infections and failure to mount
1998; Ishikawa et al., 2006; Stanley et al., 2011). IL-6, mainly protective immunity upon vaccination (Sheridan et al., 2012).
secreted by Kupffer cells (resident liver macrophages) and he- Obese individuals have fewer cytotoxic CD8+ T cells and a
patic stellate cells (HSCs), also induces IR and its effect is reduced number of NK cells with lower cytotoxic activity (Lamas
reversed by treatment with IL-6 neutralizing antibodies (Klover et al., 2002). Animal studies have corroborated these findings.
et al., 2005). Furthermore, reduced dietary intake and increased Genetically obese mice show thymic atrophy, and fewer T cells

50 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.


Cell Metabolism

Review

with reduced activation and cytotoxic capacity (Tanaka et al., 2014). Since these loci are involved in obesity, T2DM and IR,
2000). Dendritic and NK cells are also reduced (Macia et al., these findings suggest a connection between them to increased
2006). Mice on high-fat diet (HFD) recapitulate these character- HCC risk.
istics with a concomitant increase in macrophages/monocytes The physiologic hallmarks of T2DM are IR in hepatic and pe-
and similar impairment of immune responses to infection and ripheral tissues and pancreatic b-cell dysfunction (Watanabe,
immunization (Bandaru and Rajkumar, 2013). Possible mecha- 2010). Interestingly, very few T2DM susceptibility loci were
nisms driving these immune alterations could be the above- also mapped as IR loci. So where are the IR loci? Multiple inde-
mentioned systemic effects. Indeed, higher circulating amounts pendent studies show that the main IR susceptibility loci are
of pro-inflammatory cytokines can profoundly alter adaptive APOC3, PPARG, IRS1, GCKR, and IGF1 (Petersen et al.,
immune responses, although the exact mechanisms remain to 2010b; Watanabe, 2010). In 2011, a GWAS involving 1,040
be elucidated. Leptin may also be an important mediator of im- African Americans was conducted to detect genetic variation
mune dysregulation since immune cells express its receptor associated with IR from the HyperGEN study. The results high-
(Conde et al., 2014). However, Leptin exerts diverse and pleio- lighted several SNPs associated with fasting insulin and homeo-
tropic effects depending on the cell type and therefore it is stasis model assessment of IR (HOMA-IR) near the ATP10A
difficult to discern its exact role. Insulin signaling and glucose (rs6576507 and rs8026527) and CACNA1D (rs1401492) loci (Irvin
availability also affect immune cell function, as immune cell et al., 2011). In addition, the PPARg P12A GG genotype and
activation and the type of the elicited response are strongly influ- NR5A2 variants were found to be inversely associated with risk
enced by their metabolic profile (Ganeshan and Chawla, 2014). of pancreatic cancer (Tang et al., 2011).
High insulin or glucose concentrations can alter this profile and
result in a defective immune response. The direct consequence Liver Cancer
of impaired adaptive immunity in obese subjects with neoplasia Primary liver cancer is the fifth (men) and ninth (women) most
is enhanced tumor growth due to the absence of effective immu- common cancer worldwide but is the second leading cause of
nosurveillance that may retard the growth of small early tumors. cancer deaths, due to a 5 year survival rate below 10% (Ferlay
et al., 2015). Hepatocellular Carcinoma (HCC) accounts for
Genetic Factors 85%–90% of primary liver cancers (El-Serag and Rudolph,
Numerous genetic factors that affect obesity and therefore may 2007). At the time being, global HCC burden is mainly driven
potentiate obesity-related cancer risk were identified through by HBV and HCV infections, which account for 80% of cases
genome-wide association studies (GWAS) and more classical (Bosch et al., 2005), but in the near future obesity is likely to
candidate gene approaches (Grarup et al., 2014). Overall, become the leading factor, at least in the U.S. In Southeast
GWAS had successfully identified more than 100 different loci Asia, HCC incidence rates have been declining thanks to HBV
associated with increased adiposity, using BMI as a quantitative vaccination programs, but in the U.S., HCC incidence has
measure. One of the most interesting findings was identification tripled from 1977–1979 to 2005–2007 and 30%–35% of HCC pa-
of an FTO locus variant as a T2DM risk factor (Frayling et al., tients in the U.S. are virus negative (Davila and El-Serag, 2012).
2007), a finding replicated by meta-analysis and refined to Non-alcoholic fatty liver disease (NAFLD) and its more aggres-
include the MC4R locus (Loos et al., 2008). Six additional loci: sive manifestation, non-alcoholic steatohepatitis (NASH), are
TMEM18, KCTD15, GNPDA2, SH2B1, MTCH2, and NEGR1 emerging as the primary driving force behind this growth. It is
were subsequently identified (Willer et al., 2009) and shown to estimated that at least 25% of the U.S. population has NAFLD
also be of relevance for obesity in East Asians together with a (Lazo et al., 2013), with up to 8% of them exhibiting some degree
few other loci (Pei et al., 2014; Wen et al., 2012). T2D and obesity of NASH (Clark et al., 2003), with around 25% of NASH patients
were hypothesized to have an inflammatory basis, and the TNF/ progressing to cirrhosis, a serious pre-malignant condition
LTA locus has been a long-standing T2D candidate gene (Dan- (McCullough, 2004). Alarmingly, current estimates project
dona et al., 2004). However, a recent GWAS found no consistent 1.5%–2% of the U.S. population as having liver cirrhosis due
association between TNF/LTA locus variation and T2D (Boraska to NASH, and in a few years this is expected to be the leading
et al., 2010). Among all obesity risk loci, FTO has the strongest indication for liver transplantation (Holmberg et al., 2013).
effect, imposing an allelic 0.39 kg/m2 increase in BMI (Frayling Considering that the annual cumulative incidence of HCC in
et al., 2007). T2DM risk is also genetically influenced given the NASH-cirrhosis is just a bit lower than that of HCV-cirrhosis
observation that it goes up by 2- to 3-fold by having a sibling (2.6% and 4%, respectively) (Ascha et al., 2010) an upsurge of
with T2DM. Currently, about 140 genetic loci have been firmly new HCC cases is expected in the U.S. in the coming decades.
established as T2DM risk loci, with the TCF7L2 locus being the NAFLD is strongly allied to components of the metabolic syn-
strongest one. Curiously, however, despite the close pathophys- drome and most patients diagnosed with NAFLD have accom-
iological relationship between obesity and T2DM, only a small panying IR (Fabbrini et al., 2010). Due to excessive dietary intake
number of risk loci are shared between the two conditions. The as well as genetic risk factors or other diseases, accumulation
common loci are FTO, CDKAL1, GIPR, KCNQ1, LYPLAL1, of lipids in hepatocytes accompanied by de novo lipogenesis
TMEM18, ZNF608, MC4R, GRB14, RREB1, and LINGO2, but and decreased mitochondrial b oxidation could lead to liver
so far there is little or no information whether they also increase dysfunction. Mechanistically, high levels of nutrients activate
cancer risk. Obviously, such studies are needed especially in the mTOR, which in turn leads to the processing and release
case of liver and uterine cancers, where obesity constitutes a from the ER of SREBP1, the master regulator of lipogenesis,
major risk factor. Recently, IRS1, HMGCS1, ATP8B1, PRMT6, through the dissociation of CRTC2 from the COPII subunit
and CLU were found to be mutated in HCC (Meerzaman et al., (Han et al., 2015) (Figure 2). Importantly, triglycerides (TG)

Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc. 51


Cell Metabolism

Review

Figure 2. Mechanisms of Obesity-Induced HCC


Two main pathways converge to drive ER stress in the obese liver. On one hand, microbial products from the intestine reach the liver via the portal vein. Liver-
resident macrophages activate an inflammatory response that includes iNOS expression, which results in S-nitrosylation of IRE1a, thereby compromising the
UPR and consequently leading to ER stress. On the other hand, high amounts of nutrients activate mTORC1 signaling leading to phosphorylation of CRTC2,
freeing SEC31 and allowing the activation of the SEC31/32 complex that processes SREBP1. Processed SREBP1 translocates into the nucleus and activates the
lipogenic program, thereby aggravating the lipid accumulation and inducing severe hepatic steatosis with consequent lipotoxicity, ROS generation, and ER
stress. At the same time, newly synthesized lipids change the composition of the ER membrane and inhibit the sarco/endoplasmic reticulum calcium ATPase
(SERCA) activity producing a decrease of Ca2+ aggravating ER stress. Both pathways synergistically cooperate to drive hepatocyte cell death, while ROS buildup
in liver cells promotes mutagenesis and genomic instability. Importantly, cell death increases tissue inflammation and supports a positive feedback loop that
extends liver damage and compensatory proliferation. When this becomes chronic, it leads to HCC development.

accumulate in hepatocytes and cause hepatic steatosis once death of steatotic hepatocytes, which is further amplified
the storage capacity of conventional fat depots has maxed through a vicious cycle of chronic liver damage and inflamma-
out. It has been shown that free cholesterol is a potentially impor- tion. Liver damage stimulates compensatory proliferation and
tant player, sensitizing hepatocytes to TNF-induced inflamma- the main mitogens involved in this response are IL-6 and TNF,
tion after its accumulation in mitochondria (Marı́ et al., 2006). both of which play important roles in HCC development (He
Up to a point, NAFLD is a reversible condition, but without proper et al., 2010, 2013; Nakagawa et al., 2014; Naugler et al., 2007).
treatment it may progress to NASH, which is characterized by Lineage-tracing experiments in NASH-induced HCC mouse
chronic liver inflammation and ongoing tissue damage. Liver models suggest that normal centrilobular hepatocytes are the
damage results in compensatory proliferation, a major driver of cells that give rise to NASH-promoted HCC, rather than so-
HCC development (Maeda et al., 2005). called oval cells and newly identified hybrid hepatocytes, which
Since HCC almost always evolves in the context of chronic are the cells that undergo the most extensive damage-induced
liver damage, efforts have been concentrated on understanding proliferation (Font-Burgada et al., 2015). Differentiated hepato-
what drives chronic liver damage and cirrhosis in NAFLD pa- cytes are the cells that are most likely to accumulate lipids and
tients that progress to NASH and eventually HCC. The ‘‘two-hit suffer the consequences of lipid-influenced oxidative stress
hypothesis’’ suggests that, apart from lipid accumulation due (Figure 2). Importantly, most of the above-mentioned tumor-pro-
to hypernutrition and IR, an ill-defined second hit that promotes moting effects of obesity play a role in HCC development in the
oxidative stress, endoplasmic reticulum (ER) stress, inflamma- NASH-affected liver (Nakagawa et al., 2014). However, obesity-
tion, and hepatocyte cell death is necessary for progression induced IR is as pronounced in wild-type mice that develop sim-
from steatosis to NASH (Day and James, 1998). Oxidative and ple steatosis as it is in MUP-uPA mice that develop full-blown
ER stresses were proposed as the main culprits in driving the NASH, and therefore IR is not a major rate-determining factor

52 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.


Cell Metabolism

Review

in NASH development and its progression to HCC (Nakagawa instability (Aghajan et al., 2012). Systemic mosaic deletion of
et al., 2014). Nevertheless, central obesity and steatohepatitis Atg5 or liver-specific deletion of Atg7 results in liver damage
also greatly enhance carcinogen-induced HCC, and IL-6 and and the development of benign liver adenomas (Komatsu
TNF play important roles in this process (Park et al., 2010). et al., 2005; Takamura et al., 2011). Hepatic steatosis leads
Notably, in mice injected with the hepatic carcinogen DEN, die- to chronic activation of the mTORC1 kinase complex, which
tary sugar intake was found to contribute to liver tumor burden blocks the initiation of autophagy through inhibitory ULK1
independently of excess adiposity or IR (Healy et al., 2015). In phosphorylation (Kim et al., 2011) and unrestrained mTORC1
mice as in humans, the tumor-promoting effect of obesity is activation leads to HCC development in mice (Menon et al.,
much more pronounced in males than in females. Although he- 2012). Hypernutrition also inhibits AMPK, whose activation pro-
patic steatosis and de novo lipogenesis may not be directly motes the initiation of autophagy through stimulatory ULK1
involved in cancer cell growth and malignant progression, they phosphorylation (Kim et al., 2011). One way to prevent exces-
provide a more cancer-supportive microenvironment by gener- sive inhibition of autophagy during hepatic steatosis involves
ating inflammatory signals that exacerbate liver inflammation induction of sestrins, which are evolutionarily conserved
and activate resident and newly recruited macrophages that pro- AMPK activators (Lee et al., 2013). Conversely, persistent he-
duce TNF and IL-6. These cytokines promote malignant progres- patocyte AMPK deficiency due to TAK1 ablation results in auto-
sion through activation of NF-kB and STAT3 in initiated hepato- phagy defects and leads to HCC development, even in the
cytes (Nakagawa et al., 2014). In addition, lipid accumulation can absence of ongoing hepatic steatosis (Inokuchi-Shimizu et al.,
result in ER stress (Hummasti and Hotamisligil, 2010), a common 2014; Seki et al., 2009). In fact, the phenotype of Tak1Dhep
feature in many liver diseases, including viral hepatitis (Waris mice is very similar to that of Tsc1Dhep mice (Menon et al.,
et al., 2002). ER stress is characterized with an altered composi- 2012). Because autophagy also leads to degradation of lipid
tion of lipids in the ER and inhibition of the sarco/endoplasmic re- droplets, its inhibition contributes to hepatic steatosis in HFD-
ticulum calcium ATPase (SERCA) (Fu et al., 2011). The resulting fed mice (Schneider and Cuervo, 2014). Defective autophagy
elevated inflammation in turn induces iNOS activity producing also results in accumulation of p62, an autophagy substrate
nitrosylation of hepatic IRE1a, which subsequently fails to splice and chaperon that directs ubiquitinated protein aggregates
XBP1, preventing production of ER chaperons (Yang et al., 2015) and organelles to autophagosomes for subsequent lysosomal
(Figure 2). Finally, unresolved ER stress leads to oxidative stress degradation. Deletion of p62 reverses liver injury and inhibits
that promotes necrotic cell death, liver damage, inflammation, adenoma development in Atg7Dhep mice (Komatsu et al.,
and compensatory proliferation as shown in a novel mouse 2007). Notably, p62 is a component of Mallory bodies, inclusion
model of NASH-induced HCC (Nakagawa et al., 2014). TNF, bodies that are a common pathological feature of different liver
which acts as a growth factor for HCC progenitor cells, is likely diseases, including alcoholic hepatitis, NASH, and HCC (Zat-
to be an important player in human NASH, and the use of anti- loukal et al., 2002). Accumulation of p62 or insufficient auto-
TNF drugs for other morbidities, such as psoriatic arthritis phagy activates nuclear factor erythroid 2-related factor 2
(PsA), was observed to result in reduced NASH severity, as (NRF2) through titration of Kelch-like ECH-associated protein
long as it was effective in reducing PsA disease activity (Di Minno 1 (Keap1) (Komatsu et al., 2010). Despite its ability to induce
et al., 2012). Other immune cells that contribute to NASH devel- expression of anti-oxidant enzymes, NRF2 activation may
opment and malignant progression are CD8+ T cells, NKT cells, contribute to HCC development (Inami et al., 2011).
and cells that produce lymphotoxin (LT), a TNF family member Efficient autophagy plays an important role in restraining
whose blockade offers another option for interfering with inflammation. Apoptosis or necrosis can be observed upon
NASH to HCC progression (Wolf et al., 2014). Unlike TNF, how- long-term suppression of autophagy, and autophagy defects
ever, there is little information regarding the involvement of LT in promote inflammation and tumorigenesis (White et al., 2010).
human obesity and obesity-promoted cancer. Certain NOD-like receptors (NLRs), a group of pattern recogni-
Macroautophagy (hereafter referred to as autophagy) is tion receptors with diverse functions, such as NLRP3, mediate
an evolutionarily ancient proteolytic process that begins with activation of caspase-1 and secretion of mature IL-1b and IL-
formation of an autophagosome that delivers engulfed cellular 18 by activated myeloid cells (Franchi et al., 2012). Interestingly,
constituents to the lysosome, where they are degraded. NLRP3 can sense the presence of cholesterol crystals and
Autophagy-related genes (ATGs), which are evolutionarily con- various other DAMPs leading to IL-1 and IL-18 release. A poten-
served, control autophagosome formation and fusion with lyso- tial link between inflammasome activation and autophagy was
somes. In addition to providing essential building blocks during first observed when loss of the autophagy protein Atg16L1
periods of starvation, autophagy is also a quality-control mech- was found to enhance IL-1b and IL-18 production (Saitoh
anism that maintains cellular homeostasis and prevents oxida- et al., 2008). Autophagy blockade can result in accumulation of
tive stress (Yen and Klionsky, 2008). Constitutive autophagy damaged mitochondria that produce reactive oxygen species
prevents accumulation of misfolded proteins and damaged or- (mtROS) and leak mitochondrial (mt) DNA, both of which stimu-
ganelles, including ROS-producing non-functional mitochon- late NLRP3-dependent caspase-1 activity and result in IL-1b
dria and damaged ER membranes (Figure 2). In this sense, and IL-18 production (Zhou et al., 2011), further explaining how
obesity is a condition inextricably linked to defective autophagy long-term inhibition of autophagy can promote inflammation.
characterized by a marked downregulation of ATG7. Restora- By contrast, efficient autophagy can limit NF-kB activation
tion of ATG7 in the liver normalizes ER and insulin sensitivity and TNF receptor-associated factor 6 (TRAF6) signaling, both
(Yang et al., 2010). Thus, by inhibiting autophagy, hypernutrition of which are involved in inflammation-mediated tumor promo-
can cause oxidative stress, thereby promoting chromosomal tion, by enhancing the degradation of p62/SQSTM1 (Moscat

Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc. 53


Cell Metabolism

Review

and Diaz-Meco, 2012). Autophagy can also prevent secondary also confirmed differential expression of GC and LCP1 in a
necrosis and DAMP release by clearing apoptotic bodies. Both NAFLD cohort (Adams et al., 2013).
ATG5 and damage-regulated autophagy modulator (DRAM)
are required for p53-induced apoptotic death (Crighton et al., Pancreatic Cancer
2006), a tumor-suppressive mechanism. Conversely, Atg5 abla- Pancreatic ductal adenocarcinoma (PDAC) is another highly
tion results in impaired clearance of apoptotic bodies and conse- aggressive malignancy with a 5 year survival rate of 6%. PDAC
quent inflammation (Qu et al., 2007). risk is also strongly enhanced by obesity (Calle et al., 2003; Wise-
Dysbiosis has been detected in liver cirrhosis patients (Bajaj man, 2008) and PDAC is predicted to become one of the three
et al., 2014). As mentioned above, obesity induces changes top lethal cancers in the U.S. by 2030, together with lung and
in microbiome composition and gut permeability, thereby liver cancers (Rahib et al., 2014). PDAC development depends
increasing endotoxin concentration in the portal circulation, on accumulation of KRAS-activating mutations in pancreatic
which connects the gut to the liver. Portal endotoxin stimu- acinar cells, which give rise to premalignant PanIN (Pancreatic
lates production of inflammatory cytokines by both Kupffer Intraepithelial Neoplasia) lesions (Kopp et al., 2012). Curiously,
cells and newly recruited liver macrophages, thereby promot- however, low-grade PanIN harboring oncogenic KRAS alleles
ing HCC development (Schwabe and Jobin, 2013) (Figure 2). are very common in older individuals (R70 years), but the major-
The altered gut microbiota of obese mice also produces ity of these individuals never develop PDAC (Hruban et al., 2000;
higher than normal amounts of deoxycholic acid (DCA), a Lüttges et al., 1999), suggesting that additional genetic events
secondary bile acid that enhances secretion of cytokines are needed for malignant progression. In addition to old age
by senescent hepatic stellate cells (HSCs), a process that and obesity, PDAC risk factors include chronic pancreatic
was reported to promote HCC development in carcinogen- inflammation (pancreatitis), alcohol consumption, and tobacco
treated mice (Yoshimoto et al., 2013). It should be noted, smoking (Vincent et al., 2011). Chronic inflammation caused by
however, that HSC senescence is usually associated with res- pancreatitis, obesity, and tumor-associated inflammation could
olution of inflammation and fibrosis rather than active liver dis- contribute to PDAC genesis and development (Figure 3). How-
ease, thus questioning the physiological relevance of these ever, the underlying mechanisms linking obesity and inflamma-
findings. tion to PDAC have not been fully elucidated. Oncogenic KRAS
Genetic factors are also likely to contribute to obesity-pro- can activate STAT3 and NF-kB signaling by inducing production
moted HCC. NAFLD tends to cluster in families and hepatic of IL-6, TNF, and IL-11 (Ling et al., 2012). Moreover, activation of
steatosis is more common in siblings and parents of children NF-kB by TNF can stimulate Notch signaling, which synergizes
with NAFLD compared to overweight children without NAFLD with oncogenic KRAS to further promote PDAC development
(Schwimmer et al., 2009). NAFLD heritability was estimated to (Maniati et al., 2011). Obesity can also promote infiltration of
be 39% after adjusting for age, sex, race, and BMI. So far, a num- the pancreas with immunosuppressive cells, including Treg
ber of potential genetic determinants were identified by GWAS. and so-called myeloid-suppressive cells, that can prevent the
The non-synonymous rs738409 (I148M) SNP in the PNPLA3 immunosurveillance of early, malignant lesions (Cox and Olive,
gene, which encodes a triacylglycerol lipase that mediates tri- 2012). Obesity also increases macrophage infiltration into
glyceride hydrolysis in adipocytes, is the first and most important pancreatic islets (Ehses et al., 2007). Altogether, immune cells
NAFLD-associated genetic variant (Romeo et al., 2008). These account for roughly 50% of the pancreatic tumor cell mass,
findings were confirmed by another GWAS (Speliotes et al., and most of them are immunosuppressive in nature (Feig et al.,
2011) and are of significance even after adjustment for BMI, 2012). Obesity and inflammation can also promote metastasis,
T2DM, alcohol use, and ethnicity. A meta-analysis pooling the a common complication of PDAC, by stimulating epithelial-
results of 16 studies showed that PNPLA3 rs738409 also had a mesenchymal transition (EMT). Treatment with the immunosup-
strong association with a more aggressive disease, as homozy- pressive agent dexamethasone blocks metastatic dissemination
gotes had 3.5-fold greater risk of NASH, 3.3-fold greater risk of (Rhim et al., 2012).
fibrosis, and 28% increase in serum ALT, an indicator of liver Evidence from animal models indicates that dysfunctional
damage (Sookoian and Pirola, 2011). Moreover, the PNPLA3 autophagy is associated with pancreatitis and most of the
rs738409 C > G polymorphism is not only associated with PDAC risk factors mentioned above are known to interfere with
greater risk of progressive steatohepatitis and fibrosis but also either initiation or completion of autophagy (Mareninova et al.,
of HCC (Liu et al., 2014). A pilot GWAS of NAFLD, using a rela- 2009) (Figure 3). Defective lysosomal proteolysis results in for-
tively small all-female cohort of 236 patients, identified an asso- mation of large cytoplasmic vacuoles, which is a long-noted
ciation between NAFLD activity score and the rs2645424 SNP in but poorly understood pathological feature of pancreatitis (Gu-
the FDFT1 gene, which encodes an enzyme involved in choles- kovskaya and Gukovsky, 2012). Autolysosomes account for
terol biosynthesis (Chalasani et al., 2010). Several more SNPs most of the vacuoles that accumulate in acinar cells during
associated with NAFLD were found in the PNPLA3 (rs738408), pancreatitis. As indicated above, defective autophagy signifi-
NCAN (rs2228603), PPP1R3B (rs4240624), GCKR (rs780094), cantly extends the half-life of dysfunctional organelles, including
and the LYPLAL1 (rs12137855) loci (Speliotes et al., 2011). The damaged mitochondria that produce mtROS and release
rs738408 PNPLA3 SNP is in strong linkage disequilibrium with mtDNA. Decreased degradation of long-lived proteins and for-
the rs738409 SNP (Romeo et al., 2008). In addition, a recent mation of p62 aggregates also occurs during pancreatitis and
GWAS in adolescents diagnosed with NAFLD identified SNP can further exacerbate oxidative stress by triggering ER stress
associations in two liver-specific genes (GC and LCP1) and (Li et al., 2013). As discussed above, ROS accumulation leads
two neuronal-specific genes (LPPR4 and SLC38A8). This study to genomic instability and stimulates tumor initiation. Defective

54 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.


Cell Metabolism

Review

Figure 3. Mechanisms of Obesity-Promoted PDAC


Defective autophagy in pancreatic acinar cells leads to toxic accumulation of p62 aggregates, inducing ER stress and eventual cell death. Established
inflammation and NRF2 activation cause the metaplasia of surviving acinar cells and converts them to a ductal progenitor phenotype, thereby generating low-
grade PanIN. To convert low-grade PanIN lesions into PDAC, metabolic reprogramming is needed through the action of members of the MITF/TFE family of
transcription factors, which upregulate autophagy, to promote cell survival. Other metabolic changes promote proliferation within PanIN lesions and lead to
PanIN to PDAC progression.

autophagy and insufficient lysosomal proteolysis also cause the subsequently five new susceptibility loci were mapped to
pathologic, intra-acinar accumulation of trypsin, which is another 21q21.3, 5p13.1, 21q22.3, 22q13.32, and 10q26.11 (Wu et al.,
hallmark of pancreatitis (Gukovskaya and Gukovsky, 2012; Gu- 2012). The exact identity of these risk loci, their mechanism of
kovsky and Gukovskaya, 2015). In contrast to high-grade PanIN action, and whether they are related to obesity or T2DM is un-
in which autophagy is upregulated, lower-grade PanIN exhibit known.
reduced autophagy rate, and insufficient autophagy can pro-
mote the progression of early PanIN lesions to high-grade PanIN Colorectal Cancer
and PDAC. Exactly how autophagy is upregulated during later Colorectal cancer (CRC) is the third most common cancer in men
stages of malignant progression is not entirely clear, but a recent and the second in women (Ferlay et al., 2015). The sequential ge-
publication suggests that PDAC metabolic reprogramming re- netic progression of this malignancy is well established. Colonic
quires the constitutive activation of MITF, TFE3, and TFEB tran- adenomas are considered as precursors to carcinoma and their
scription factors that are otherwise retained in the cytoplasm. prevalence in the general population is strikingly high. As in
These transcription factors upregulate transcription of genes PDAC, the adenoma to carcinoma transition is highly influenced
related to lysosome biogenesis and autophagy to maintain by environmental factors. There are several explanations for the
essential amino acid pools (Perera et al., 2015) (Figure 3). high incidence of gastrointestinal (GI) cancers. First, the GI
Epidemiologic studies have identified T2DM as another epithelium contains a much larger number of actively dividing
pancreatic cancer risk factor (Lin et al., 2011). The first pancre- stem and progenitor cells than other epithelial tissues, including
atic cancer GWAS identified a common risk variant, rs505922, the liver and pancreas, where cell division is mostly evoked in
that maps to the first intron of the ABO gene (Amundadottir response to damage. Second, the GI epithelium is in direct con-
et al., 2009). This finding implies that people with blood group tact with the environment, including ingested material that may
O may have a lower risk than those with groups A or B. A second contain trace amounts of toxins and carcinogens. Third, the GI
GWAS identified additional pancreatic cancer susceptibility loci tract, especially the colon, is a host to trillions of commensal mi-
at 13q22.1, 1q32.1, and 5p15.33 (Petersen et al., 2010a) and crobes. Although these microbes are normally well-tolerated,

Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc. 55


Cell Metabolism

Review

Figure 4. Mechanisms of Obesity-Induced CRC


Obesity induces dysbiosis and thinning of the intestinal mucus layer resulting in increased permeability of the intestinal epithelium to microbial products. Resident
immune cells react to these inflammatory cues by secreting a cocktail of inflammatory cytokines that act directly on cancer progenitor cells to stimulate their
survival and proliferation. Obesity-associated microbial dysbiosis can result in elevated concentrations of metabolites that further contribute to tumor promotion.
High levels of circulating IGF-1, insulin and inflammatory cytokines, and reduced levels of adiponectin also contribute to promotion of CRC development.

certain diets, including HFD, can lead to dysbiosis and thereby results in similar outcomes (Moon et al., 2013). Mouse studies
trigger an inflammatory response. Fourth, the GI epithelium have also shown that leptin acts as a growth factor in CRC
serves as a barrier that prevents penetration of luminal microbes (Endo et al., 2011) (Figure 4). Obesity and hyperinsulinemia
and their disintegration products (endotoxin, nucleic acids, etc.) also increase expression of IGF-1, another well-established fac-
into the underlying tissue, where they can evoke a tumor-pro- tor that promotes CRC development and malignant progression
moting inflammatory response (Grivennikov et al., 2012). In addi- by binding IGFR. Obesity-induced alterations in intestinal barrier
tion to HFD, diets that are fiber poor also reduce microbiome permeability have an additional influence on CRC development.
diversity and enhance bowel inflammation (Poullis et al., 2004). HFD-fed mice show increased circulating LPS and such endo-
Altogether, such diets play on a common theme in cancer devel- toxemia can promote CRC development (Cani et al., 2007).
opment: increased tissue damage and consequent tumor-pro- Monocolonization of germ-free (GF) mice with different E. coli
moting inflammation (Grivennikov et al., 2010) (Figure 4). strains that produce distinct forms of LPS showed that only
The risk and incidence of CRC go up with increased BMI (Calle mice exposed to immunogenic LPS exhibited accumulation of
et al., 2003). Meta-analysis revealed a 5% increase in CRC inci- pro-inflammatory macrophages in adipose tissue (Caesar
dence per inch of above-normal waist circumference (Wiseman, et al., 2012). These results suggest that obesity-induced barrier
2008). HFD-fed mice showed a marked increase in colonic TNF disruption results in mild endotoxemia that synergizes with
expression (Liu et al., 2012). TNF is a potent inducer of IL-6, pre-existing adipose tissue inflammation to further increase tu-
whose role in promoting CRC has been widely studied and mor-promoting systemic inflammation. In addition to TNF and
recently reviewed (Taniguchi and Karin, 2014). By contrast, adi- IL-6, IL-22 was shown to play a central role in establishing meta-
ponectin can inhibit the growth of CRC cells by activating AMPK bolic imbalances observed in obesity. HFD-fed IL-22R-deficient
(Sugiyama et al., 2009) and, consequently, low adiponectin mice showed increased propensity to develop metabolic disor-
levels are associated with increased CRC risk (Gialamas et al., ders, and administration of IL-22 into genetically obese db/db
2011; Otake et al., 2010). Furthermore, adiponectin ablation en- mice or mice fed with HFD resulted in a significant amelioration
hances development of colitis-associated cancer or intestinal of IR, hyperglycemia, intestinal barrier disruption, endotoxemia,
adenomas in Apcmin mice (Mutoh et al., 2011) and HFD feeding and systemic inflammation (Wang et al., 2014). Despite the fact

56 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.


Cell Metabolism

Review

that IL-22 can have initial anti-tumorigenic activity, its signaling the most cost-effective way to reduce the enormous cancer-
can be corrupted by malignant cells to enhance survival and related burden of overweight and obesity (Calle et al., 2003).
metastasis (Lim and Savan, 2014), thus obscuring its therapeutic NSAIDs and anti-TNF drugs may also act synergistically with
potential. other treatments. It was reported that PsA patients receiving
Notably, a recent paper had suggested that the pro-tumori- anti-TNF drugs presented less hepatostetaosis (Di Minno et al.,
genic effect on HFD on intestinal cancer is not related to obesity 2012). Similar observations were made on the degree of IR in
per se and that tumor promotion is mainly due to dysbiosis. Fecal RA patients on anti-TNF therapy (Burska et al., 2015; Solomon
transfer from HFD-fed Kras(G12Dint) mice was sufficient to et al., 2011). The antidiabetic drug metformin has shown prom-
enhance tumorigenic growth and progression in the absence ising effects in reducing cancer-associated risk in diabetic pa-
of HFD in recipient mice (Schulz et al., 2014) In addition, antibi- tients (Evans et al., 2005; Quinn et al., 2013) and some positive ef-
otic treatment blunted the HFD-induced tumor progression. fects in NAFLD (Marchesini et al., 2001), although not in NASH.
Metformin’s anti-cancer effects appear to depend on mTORC1
Prevention, Risk Prediction, and Cancer Treatment inhibition, specifically in cancer stem cells (Hirsch et al., 2009).
Importantly and despite its profound impact, much of the effect However, long-term mTORC1 inhibition has adverse effects on
of obesity on cancer development and mortality can be pre- the liver and may increase HCC risk (Umemura et al., 2014).
vented or even reversed by dietary modification, increased ex- More potent and specific interventions, however, may be
ercise regimens, or, in more severe cases, bariatric surgery. needed for NASH-driven HCC. First, unlike the low-grade inflam-
Since obesity and hypernutrition mainly act as tumor pro- mation associated with obesity, NASH is a serious inflammatory
moters, even delayed intervention, after early malignancy has disease that can frequently lead to liver failure well before
been detected, may be beneficial, if not by itself, then in com- appearance of HCC. Mouse studies show that TNF and the
bination with chemo- or immunotherapy. Early intervention related cytokine LT play important roles not only in the develop-
together with administration of potent insulin-sensitizing drugs ment of NASH-related HCC but also in the pathogenesis of
and IGF antagonists may prevent the development of all NASH itself (Nakagawa et al., 2014; Wolf et al., 2014). Curiously,
obesity-promoted cancers, leading to a 15% decrease in LT also regulates the ability of commensal bacteria to promote
cancer-related deaths (Calle et al., 2003). However, given the weight gain (Upadhyay et al., 2012). Thus, LT antagonists
failure of behavioral modification and bariatric surgery in stem- together with established anti-TNF drugs may be of particular
ming the impact of the obesity epidemic until now, more so- therapeutic value in this obesity-induced cancer. The chemical
phisticated pharmacological and therapeutic approaches are chaperon TUDCA has shown to reduce inflammation in the adi-
needed. These may include PPARg modulation, induction of pose tissue in obese mice (Kawasaki et al., 2012) as well as to be
brown fat via drug treatment or cell transplantation, inhibition effective in normalizing fatty liver disease in mice and improving
of adipose tissue angiogenesis, or adipokine therapy, although insulin sensitivity in mice and humans (Kars et al., 2010; Ozcan
in most cases the validity and feasibility of such approaches re- et al., 2006). Although TUDCA had shown efficacy in mouse
mains to be demonstrated (Rosen and Spiegelman, 2014). models of NASH (Nakagawa et al., 2014), it has not produced
Another plausible, but not yet tested, approach is restoration remarkable effects at normal doses in human NASH. Vitamin
of microbiome diversity in those cases where obesity or hyper- E, however, has shown promise in the treatment of human
nutrition lead to pronounced dysbiosis. As a matter of fact, it NASH. The protective and therapeutic effects of vitamin E may
was recently reported that bariatric surgery elicits sustained be related to its antibiotic activity.
changes in microbiome composition along with weight loss Obesity not only increases cancer risk but also increases its
that are transmissible to recipient germ-free mice upon fecal mortality rate (Calle et al., 2003). Little is known about the
transfer (Tremaroli et al., 2015). intrinsic characteristics of tumors promoted by obesity. For
As discussed above, common genetic variants impose modest instance, it is not yet known whether obesity affects the muta-
risk increments on various parameters of the metabolic syn- tional spectrum associated with tumor initiation and progression.
drome and associated cancers. So far, GWASs have not resulted Such knowledge is of particular importance in the case of the
in identification of obvious therapeutic targets and it is not clear most lethal obesity-promoted cancers reviewed above. For
whether addressing a factor that increases relative disease risk instance, a specific oncogenic driver induced by obesity may
by only a small increment is a worthwhile effort. It is much clearer offer a specific and unique target for drug development, in addi-
that regardless of its initial tissue-specific effects, obesity in- tion to the more general approaches discussed above.
creases cancer risk and accelerates malignant progression
by causing low-grade chronic inflammation. Thus, anti-inflam- Conclusions
matory intervention may be the most effective preventative
approach for the majority of obesity-promoted cancers. Indeed,
there is considerable epidemiological evidence that long-term d Obesity increases the risk of developing deadly malig-
consumption of Aspirin and other non-steroidal anti-inflamma- nancies through a panoply of pathophysiological alter-
tory drugs (NSAID) results in a considerable decrease in cancer ations including systemic inflammation, dysregulation of
risk, as first identified in CRC (Flossmann et al., 2007), but now adipokines, insulin resistance with hyperinsulinemia and
extended to several other cancers as well (Rothwell et al., hyperglycemia, dysbiosis, and immune system alterations.
2011). Importantly, Aspirin and other salicilates were also proven d Obesity-promoted cancers are amongst the most deadly
effective in the treatment of IR and other aspects of the metabolic and are predicted to become leading killers by 2030 in
syndrome (Goldfine et al., 2013). Thus, an Aspirin a day may be the U.S.

Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc. 57


Cell Metabolism

Review
d The key to tackling these cancers is to understand the Calle, E.E., and Kaaks, R. (2004). Overweight, obesity and cancer: epidemio-
mechanisms underlying increased risk, thereby identifying logical evidence and proposed mechanisms. Nat. Rev. Cancer 4, 579–591.
patients who are at highest risk of developing obesity-pro- Calle, E.E., Rodriguez, C., Walker-Thurmond, K., and Thun, M.J. (2003). Over-
moted malignancy. Efforts should be directed to under- weight, obesity, and mortality from cancer in a prospectively studied cohort of
U.S. adults. N. Engl. J. Med. 348, 1625–1638.
standing the progression of NAFLD to NASH and that of
NASH to HCC and at development of effective early pre- Cani, P.D., Amar, J., Iglesias, M.A., Poggi, M., Knauf, C., Bastelica, D., Neyr-
vention for high-risk individuals. inck, A.M., Fava, F., Tuohy, K.M., Chabo, C., et al. (2007). Metabolic endotox-
emia initiates obesity and insulin resistance. Diabetes 56, 1761–1772.
d The human microbiota was proposed as an important
player in establishment of pathophysiological conditions Catrina, S.B., Okamoto, K., Pereira, T., Brismar, K., and Poellinger, L. (2004).
associated with obesity and obesity-induced cancer. A Hyperglycemia regulates hypoxia-inducible factor-1alpha protein stability
and function. Diabetes 53, 3226–3232.
stronger mechanistic understanding of effects attributed
to dysbiosis and human trials of microbiota restoration Chalasani, N., Guo, X., Loomba, R., Goodarzi, M.O., Haritunians, T., Kwon, S.,
Cui, J., Taylor, K.D., Wilson, L., Cummings, O.W., et al.; Nonalcoholic Steato-
are needed to validate the relevance and utility of this hepatitis Clinical Research Network (2010). Genome-wide association study
approach. identifies variants associated with histologic features of nonalcoholic Fatty
d Given the fundamental role of inflammation in obesity pro- liver disease. Gastroenterology 139, 1567–1576, 1576.e1–1576.e6.
moted malignancy, mainly benign anti-inflammatory treat- Chassaing, B., Koren, O., Goodrich, J.K., Poole, A.C., Srinivasan, S., Ley, R.E.,
ments should be given a priority to be evaluated as general and Gewirtz, A.T. (2015). Dietary emulsifiers impact the mouse gut microbiota
and inexpensive preventive strategies. promoting colitis and metabolic syndrome. Nature 519, 92–96.

Cheung, A.T., Ree, D., Kolls, J.K., Fuselier, J., Coy, D.H., and Bryer-Ash, M.
(1998). An in vivo model for elucidation of the mechanism of tumor necrosis
REFERENCES factor-alpha (TNF-alpha)-induced insulin resistance: evidence for differential
regulation of insulin signaling by TNF-alpha. Endocrinology 139, 4928–4935.
Adams, L.A., White, S.W., Marsh, J.A., Lye, S.J., Connor, K.L., Maganga, R.,
Ayonrinde, O.T., Olynyk, J.K., Mori, T.A., Beilin, L.J., et al. (2013). Association Cho, I., and Blaser, M.J. (2012). The human microbiome: at the interface of
between liver-specific gene polymorphisms and their expression levels with health and disease. Nat. Rev. Genet. 13, 260–270.
nonalcoholic fatty liver disease. Hepatology 57, 590–600.
Clark, J.M., Brancati, F.L., and Diehl, A.M. (2003). The prevalence and etiology
Aghajan, M., Li, N., and Karin, M. (2012). Obesity, autophagy and the patho- of elevated aminotransferase levels in the United States. Am. J. Gastroenterol.
genesis of liver and pancreatic cancers. J. Gastroenterol. Hepatol. 27 (Suppl 98, 960–967.
2 ), 10–14.
Conde, J., Scotece, M., Abella, V., López, V., Pino, J., Gómez-Reino, J.J., and
Aguirre, V., Werner, E.D., Giraud, J., Lee, Y.H., Shoelson, S.E., and White, M.F. Gualillo, O. (2014). An update on leptin as immunomodulator. Expert Rev. Clin.
(2002). Phosphorylation of Ser307 in insulin receptor substrate-1 blocks inter- Immunol. 10, 1165–1170.
actions with the insulin receptor and inhibits insulin action. J. Biol. Chem. 277,
1531–1537. Considine, R.V., Sinha, M.K., Heiman, M.L., Kriauciunas, A., Stephens, T.W.,
Nyce, M.R., Ohannesian, J.P., Marco, C.C., McKee, L.J., Bauer, T.L., et al.
Amundadottir, L., Kraft, P., Stolzenberg-Solomon, R.Z., Fuchs, C.S., Petersen, (1996). Serum immunoreactive-leptin concentrations in normal-weight and
G.M., Arslan, A.A., Bueno-de-Mesquita, H.B., Gross, M., Helzlsouer, K., Ja- obese humans. N. Engl. J. Med. 334, 292–295.
cobs, E.J., et al. (2009). Genome-wide association study identifies variants
in the ABO locus associated with susceptibility to pancreatic cancer. Nat. Cox, A.D., and Olive, K.P. (2012). Silencing the killers: paracrine immune sup-
Genet. 41, 986–990. pression in pancreatic cancer. Cancer Cell 21, 715–716.

Antuna-Puente, B., Feve, B., Fellahi, S., and Bastard, J.P. (2008). Adipokines: Crighton, D., Wilkinson, S., O’Prey, J., Syed, N., Smith, P., Harrison, P.R.,
the missing link between insulin resistance and obesity. Diabetes Metab. 34, Gasco, M., Garrone, O., Crook, T., and Ryan, K.M. (2006). DRAM, a p53-
2–11. induced modulator of autophagy, is critical for apoptosis. Cell 126, 121–134.
Ascha, M.S., Hanouneh, I.A., Lopez, R., Tamimi, T.A., Feldstein, A.F., and Zein, Dalamaga, M., Diakopoulos, K.N., and Mantzoros, C.S. (2012). The role of adi-
N.N. (2010). The incidence and risk factors of hepatocellular carcinoma in pa- ponectin in cancer: a review of current evidence. Endocr. Rev. 33, 547–594.
tients with nonalcoholic steatohepatitis. Hepatology 51, 1972–1978.
Dandona, P., Aljada, A., and Bandyopadhyay, A. (2004). Inflammation: the link
Bajaj, J.S., Heuman, D.M., Hylemon, P.B., Sanyal, A.J., White, M.B., Monteith, between insulin resistance, obesity and diabetes. Trends Immunol. 25, 4–7.
P., Noble, N.A., Unser, A.B., Daita, K., Fisher, A.R., et al. (2014). Altered profile
of human gut microbiome is associated with cirrhosis and its complications. Davila, J.A., and El-Serag, H. (2012). The rising incidence of hepatocellular car-
J. Hepatol. 60, 940–947. cinoma in the United States: an update. Gastroenterology 142.
Bandaru, P., Rajkumar, H., and Nappanveettil, G. (2013). The impact of obesity
Day, C.P., and James, O.F. (1998). Steatohepatitis: a tale of two ‘‘hits’’?
on immune response to infection and vaccine: an insight into plausible mech-
Gastroenterology 114, 842–845.
anisms. Endocrinol. Metab. Syndr. 2, 113.

Boraska, V., Rayner, N.W., Groves, C.J., Frayling, T.M., Diakite, M., Rockett, Di Minno, M.N., Iervolino, S., Peluso, R., Russolillo, A., Lupoli, R., Scarpa, R., Di
K.A., Kwiatkowski, D.P., Day-Williams, A.G., McCarthy, M.I., and Zeggini, E. Minno, G., and Tarantino, G.; CaRRDS Study Group (2012). Hepatic steatosis
(2010). Large-scale association analysis of TNF/LTA gene region polymor- and disease activity in subjects with psoriatic arthritis receiving tumor necrosis
phisms in type 2 diabetes. BMC Med. Genet. 11, 69. factor-a blockers. J. Rheumatol. 39, 1042–1046.

Bosch, F.X., Ribes, J., Cléries, R., and Dı́az, M. (2005). Epidemiology of hepa- Ehses, J.A., Perren, A., Eppler, E., Ribaux, P., Pospisilik, J.A., Maor-Cahn, R.,
tocellular carcinoma. Clin. Liver Dis. 9, 191–211, v. Gueripel, X., Ellingsgaard, H., Schneider, M.K., Biollaz, G., et al. (2007).
Increased number of islet-associated macrophages in type 2 diabetes. Dia-
Burska, A.N., Sakthiswary, R., and Sattar, N. (2015). Effects of Tumour Necro- betes 56, 2356–2370.
sis Factor Antagonists on Insulin Sensitivity/Resistance in Rheumatoid
Arthritis: A Systematic Review and Meta-Analysis. PLoS ONE 10, e0128889. El-Haschimi, K., Pierroz, D.D., Hileman, S.M., Bjørbaek, C., and Flier, J.S.
(2000). Two defects contribute to hypothalamic leptin resistance in mice
Caesar, R., Reigstad, C.S., Bäckhed, H.K., Reinhardt, C., Ketonen, M., Lun- with diet-induced obesity. J. Clin. Invest. 105, 1827–1832.
dén, G.O., Cani, P.D., and Bäckhed, F. (2012). Gut-derived lipopolysaccharide
augments adipose macrophage accumulation but is not essential for impaired El-Serag, H.B., and Rudolph, K.L. (2007). Hepatocellular carcinoma: epidemi-
glucose or insulin tolerance in mice. Gut 61, 1701–1707. ology and molecular carcinogenesis. Gastroenterology 132, 2557–2576.

58 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.


Cell Metabolism

Review
Ellulu, M., Abed, Y., Rahmat, A., Ranneh, Y., and Ali, F. (2014). Epidemiology of Grivennikov, S.I., Wang, K., Mucida, D., Stewart, C.A., Schnabl, B., Jauch, D.,
obesity in developing countries: challenges and prevention. Global Epidemic Taniguchi, K., Yu, G.Y., Osterreicher, C.H., Hung, K.E., et al. (2012). Adenoma-
Obesity 2. linked barrier defects and microbial products drive IL-23/IL-17-mediated
tumour growth. Nature 491, 254–258.
Endo, H., Hosono, K., Uchiyama, T., Sakai, E., Sugiyama, M., Takahashi, H.,
Nakajima, N., Wada, K., Takeda, K., Nakagama, H., and Nakajima, A. (2011). Gukovskaya, A.S., and Gukovsky, I. (2012). Autophagy and pancreatitis. Am.
Leptin acts as a growth factor for colorectal tumours at stages subsequent J. Physiol. Gastrointest. Liver Physiol. 303, G993–G1003.
to tumour initiation in murine colon carcinogenesis. Gut 60, 1363–1371.
Gukovsky, I., and Gukovskaya, A.S. (2015). Impaired autophagy triggers
Evans, J.M., Donnelly, L.A., Emslie-Smith, A.M., Alessi, D.R., and Morris, A.D. chronic pancreatitis: lessons from pancreas-specific atg5 knockout mice.
(2005). Metformin and reduced risk of cancer in diabetic patients. BMJ 330, Gastroenterology 148, 501–505.
1304–1305.
Han, J., Li, E., Chen, L., Zhang, Y., Wei, F., Liu, J., Deng, H., and Wang, Y.
Fabbrini, E., Sullivan, S., and Klein, S. (2010). Obesity and nonalcoholic fatty (2015). The CREB coactivator CRTC2 controls hepatic lipid metabolism by
liver disease: biochemical, metabolic, and clinical implications. Hepatology regulating SREBP1. Nature 524, 243–246.
51, 679–689.
Haslam, D.W., and James, W.P. (2005). Obesity. Lancet 366, 1197–1209.
Fasshauer, M., and Blüher, M. (2015). Adipokines in health and disease.
He, G., Yu, G.Y., Temkin, V., Ogata, H., Kuntzen, C., Sakurai, T., Sieghart, W.,
Trends Pharmacol. Sci. 36, 461–470.
Peck-Radosavljevic, M., Leffert, H.L., and Karin, M. (2010). Hepatocyte IKK-
beta/NF-kappaB inhibits tumor promotion and progression by preventing
Feig, C., Gopinathan, A., Neesse, A., Chan, D.S., Cook, N., and Tuveson, D.A.
oxidative stress-driven STAT3 activation. Cancer Cell 17, 286–297.
(2012). The pancreas cancer microenvironment. Clin. Cancer Res. 18, 4266–
4276. He, G., Dhar, D., Nakagawa, H., Font-Burgada, J., Ogata, H., Jiang, Y., Shala-
pour, S., Seki, E., Yost, S.E., Jepsen, K., et al. (2013). Identification of liver can-
Ferlay, J., Soerjomataram, I., Dikshit, R., Eser, S., Mathers, C., Rebelo, M., cer progenitors whose malignant progression depends on autocrine IL-6
Parkin, D.M., Forman, D., and Bray, F. (2015). Cancer incidence and mortality signaling. Cell 155, 384–396.
worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J.
Cancer 136, E359–E386. Healy, M.E., Chow, J.D., Byrne, F.L., Breen, D.S., Leitinger, N., Li, C., Lackner,
C., Caldwell, S.H., and Hoehn, K.L. (2015). Dietary effects on liver tumor
Feuerer, M., Herrero, L., Cipolletta, D., Naaz, A., Wong, J., Nayer, A., Lee, J., burden in mice treated with the hepatocellular carcinogen diethylnitrosamine.
Goldfine, A.B., Benoist, C., Shoelson, S., and Mathis, D. (2009). Lean, but J. Hepatol. 62, 599–606.
not obese, fat is enriched for a unique population of regulatory T cells that
affect metabolic parameters. Nat. Med. 15, 930–939. Hirosumi, J., Tuncman, G., Chang, L., Görgün, C.Z., Uysal, K.T., Maeda, K.,
Karin, M., and Hotamisligil, G.S. (2002). A central role for JNK in obesity and
Flossmann, E., and Rothwell, P.M.; British Doctors Aspirin Trial and the UK-TIA insulin resistance. Nature 420, 333–336.
Aspirin Trial (2007). Effect of aspirin on long-term risk of colorectal cancer:
consistent evidence from randomised and observational studies. Lancet Hirsch, H.A., Iliopoulos, D., Tsichlis, P.N., and Struhl, K. (2009). Metformin
369, 1603–1613. selectively targets cancer stem cells, and acts together with chemotherapy
to block tumor growth and prolong remission. Cancer Res. 69, 7507–7511.
Font-Burgada, J., Shalapour, S., Ramaswamy, S., Hsueh, B., Rossell, D.,
Umemura, A., Taniguchi, K., Nakagawa, H., Valasek, M.A., Ye, L., et al. Holmberg, S.D., Spradling, P.R., Moorman, A.C., and Denniston, M.M. (2013).
(2015). Hybrid Periportal Hepatocytes Regenerate the Injured Liver without Hepatitis C in the United States. N. Engl. J. Med. 368, 1859–1861.
Giving Rise to Cancer. Cell 162, 766–779.
Holzer, R.G., Park, E.J., Li, N., Tran, H., Chen, M., Choi, C., Solinas, G., and
Franchi, L., Muñoz-Planillo, R., and Núñez, G. (2012). Sensing and reacting to Karin, M. (2011). Saturated fatty acids induce c-Src clustering within mem-
microbes through the inflammasomes. Nat. Immunol. 13, 325–332. brane subdomains, leading to JNK activation. Cell 147, 173–184.

Frayling, T.M., Timpson, N.J., Weedon, M.N., Zeggini, E., Freathy, R.M., Lindg- Hotamisligil, G.S., Shargill, N.S., and Spiegelman, B.M. (1993). Adipose
ren, C.M., Perry, J.R., Elliott, K.S., Lango, H., Rayner, N.W., et al. (2007). A expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin
common variant in the FTO gene is associated with body mass index and pre- resistance. Science 259, 87–91.
disposes to childhood and adult obesity. Science 316, 889–894.
Hruban, R.H., Wilentz, R.E., and Kern, S.E. (2000). Genetic progression in the
Fu, S., Yang, L., Li, P., Hofmann, O., Dicker, L., Hide, W., Lin, X., Watkins, S.M., pancreatic ducts. Am. J. Pathol. 156, 1821–1825.
Ivanov, A.R., and Hotamisligil, G.S. (2011). Aberrant lipid metabolism disrupts
calcium homeostasis causing liver endoplasmic reticulum stress in obesity. Hummasti, S., and Hotamisligil, G.S. (2010). Endoplasmic reticulum stress and
Nature 473, 528–531. inflammation in obesity and diabetes. Circ. Res. 107, 579–591.

Inami, Y., Waguri, S., Sakamoto, A., Kouno, T., Nakada, K., Hino, O., Wata-
Gallistl, S., Sudi, K.M., Aigner, R., and Borkenstein, M. (2001). Changes in
nabe, S., Ando, J., Iwadate, M., Yamamoto, M., et al. (2011). Persistent activa-
serum interleukin-6 concentrations in obese children and adolescents during
tion of Nrf2 through p62 in hepatocellular carcinoma cells. J. Cell Biol. 193,
a weight reduction program. Int. J. Obes. Relat. Metab. Disord. 25, 1640–1643.
275–284.
Ganeshan, K., and Chawla, A. (2014). Metabolic regulation of immune re- Inokuchi-Shimizu, S., Park, E.J., Roh, Y.S., Yang, L., Zhang, B., Song, J.,
sponses. Annu. Rev. Immunol. 32, 609–634. Liang, S., Pimienta, M., Taniguchi, K., Wu, X., et al. (2014). TAK1-mediated
autophagy and fatty acid oxidation prevent hepatosteatosis and tumorigen-
Gialamas, S.P., Petridou, E.T., Tseleni-Balafouta, S., Spyridopoulos, T.N., esis. J. Clin. Invest. 124, 3566–3578.
Matsoukis, I.L., Kondi-Pafiti, A., Zografos, G., and Mantzoros, C.S. (2011).
Serum adiponectin levels and tissue expression of adiponectin receptors are Irvin, M.R., Wineinger, N.E., Rice, T.K., Pajewski, N.M., Kabagambe, E.K., Gu,
associated with risk, stage, and grade of colorectal cancer. Metabolism 60, C.C., Pankow, J., North, K.E., Wilk, J.B., Freedman, B.I., et al. (2011). Genome-
1530–1538. wide detection of allele specific copy number variation associated with insulin
resistance in African Americans from the HyperGEN study. PLoS ONE 6,
Goldfine, A.B., Fonseca, V., Jablonski, K.A., Chen, Y.D., Tipton, L., Staten, e24052.
M.A., and Shoelson, S.E.; Targeting Inflammation Using Salsalate in Type 2
Diabetes Study Team (2013). Salicylate (salsalate) in patients with type 2 dia- Ishikawa, K., Takahashi, K., Bujo, H., Hashimoto, N., Yagui, K., and Saito, Y.
betes: a randomized trial. Ann. Intern. Med. 159, 1–12. (2006). Subcutaneous fat modulates insulin sensitivity in mice by regulating
TNF-alpha expression in visceral fat. Horm. Metab. Res. 38, 631–638.
Grarup, N., Sandholt, C.H., Hansen, T., and Pedersen, O. (2014). Genetic sus-
ceptibility to type 2 diabetes and obesity: from genome-wide association Karin, M., and Lin, A. (2002). NF-kappaB at the crossroads of life and death.
studies to rare variants and beyond. Diabetologia 57, 1528–1541. Nat. Immunol. 3, 221–227.

Grivennikov, S.I., Greten, F.R., and Karin, M. (2010). Immunity, inflammation, Kars, M., Yang, L., Gregor, M.F., Mohammed, B.S., Pietka, T.A., Finck, B.N.,
and cancer. Cell 140, 883–899. Patterson, B.W., Horton, J.D., Mittendorfer, B., Hotamisligil, G.S., and Klein,

Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc. 59


Cell Metabolism

Review
S. (2010). Tauroursodeoxycholic Acid may improve liver and muscle but not Control Consortium; FUSION (2008). Common variants near MC4R are asso-
adipose tissue insulin sensitivity in obese men and women. Diabetes 59, ciated with fat mass, weight and risk of obesity. Nat. Genet. 40, 768–775.
1899–1905.
Louis, P., Hold, G.L., and Flint, H.J. (2014). The gut microbiota, bacterial me-
Kawasaki, N., Asada, R., Saito, A., Kanemoto, S., and Imaizumi, K. (2012). tabolites and colorectal cancer. Nat. Rev. Microbiol. 12, 661–672.
Obesity-induced endoplasmic reticulum stress causes chronic inflammation
in adipose tissue. Sci. Rep. 2, 799. Lüttges, J., Reinecke-Lüthge, A., Möllmann, B., Menke, M.A., Clemens, A.,
Klimpfinger, M., Sipos, B., and Klöppel, G. (1999). Duct changes and K-ras
Kim, S., Takahashi, H., Lin, W.W., Descargues, P., Grivennikov, S., Kim, Y., mutations in the disease-free pancreas: analysis of type, age relation and
Luo, J.L., and Karin, M. (2009). Carcinoma-produced factors activate myeloid spatial distribution. Virchows Arch. 435, 461–468.
cells through TLR2 to stimulate metastasis. Nature 457, 102–106.
Macia, L., Delacre, M., Abboud, G., Ouk, T.S., Delanoye, A., Verwaerde, C.,
Kim, J., Kundu, M., Viollet, B., and Guan, K.L. (2011). AMPK and mTOR regu- Saule, P., and Wolowczuk, I. (2006). Impairment of dendritic cell functionality
late autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13, and steady-state number in obese mice. J. Immunol. 177, 5997–6006.
132–141.
Maeda, S., Kamata, H., Luo, J.L., Leffert, H., and Karin, M. (2005). IKKbeta
Klover, P.J., Clementi, A.H., and Mooney, R.A. (2005). Interleukin-6 depletion couples hepatocyte death to cytokine-driven compensatory proliferation
selectively improves hepatic insulin action in obesity. Endocrinology 146, that promotes chemical hepatocarcinogenesis. Cell 121, 977–990.
3417–3427.
Maniati, E., Bossard, M., Cook, N., Candido, J.B., Emami-Shahri, N., Nedo-
Komatsu, M., Waguri, S., Ueno, T., Iwata, J., Murata, S., Tanida, I., Ezaki, J., spasov, S.A., Balkwill, F.R., Tuveson, D.A., and Hagemann, T. (2011). Cross-
Mizushima, N., Ohsumi, Y., Uchiyama, Y., et al. (2005). Impairment of starva- talk between the canonical NF-kB and Notch signaling pathways inhibits
tion-induced and constitutive autophagy in Atg7-deficient mice. J. Cell Biol. Pparg expression and promotes pancreatic cancer progression in mice.
169, 425–434. J. Clin. Invest. 121, 4685–4699.

Komatsu, M., Waguri, S., Koike, M., Sou, Y.S., Ueno, T., Hara, T., Mizushima, Marchesini, G., Brizi, M., Bianchi, G., Tomassetti, S., Zoli, M., and Melchionda,
N., Iwata, J., Ezaki, J., Murata, S., et al. (2007). Homeostatic levels of p62 con- N. (2001). Metformin in non-alcoholic steatohepatitis. Lancet 358, 893–894.
trol cytoplasmic inclusion body formation in autophagy-deficient mice. Cell
131, 1149–1163. Mareninova, O.A., Hermann, K., French, S.W., O’Konski, M.S., Pandol, S.J.,
Webster, P., Erickson, A.H., Katunuma, N., Gorelick, F.S., Gukovsky, I., and
Komatsu, M., Kurokawa, H., Waguri, S., Taguchi, K., Kobayashi, A., Ichimura, Gukovskaya, A.S. (2009). Impaired autophagic flux mediates acinar cell vacu-
Y., Sou, Y.S., Ueno, I., Sakamoto, A., Tong, K.I., et al. (2010). The selective ole formation and trypsinogen activation in rodent models of acute pancrea-
autophagy substrate p62 activates the stress responsive transcription factor titis. J. Clin. Invest. 119, 3340–3355.
Nrf2 through inactivation of Keap1. Nat. Cell Biol. 12, 213–223.
Marı́, M., Caballero, F., Colell, A., Morales, A., Caballeria, J., Fernandez, A.,
Kopp, J.L., von Figura, G., Mayes, E., Liu, F.F., Dubois, C.L., Morris, J.P., 4th, Enrich, C., Fernandez-Checa, J.C., and Garcı́a-Ruiz, C. (2006). Mitochondrial
Pan, F.C., Akiyama, H., Wright, C.V., Jensen, K., et al. (2012). Identification of free cholesterol loading sensitizes to TNF- and Fas-mediated steatohepatitis.
Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism Cell Metab. 4, 185–198.
for initiation of pancreatic ductal adenocarcinoma. Cancer Cell 22, 737–750.
McCullough, A.J. (2004). The clinical features, diagnosis and natural history of
Lamas, O., Marti, A., and Martı́nez, J.A. (2002). Obesity and immunocompe- nonalcoholic fatty liver disease. Clin. Liver Dis. 8, 521–533, viii.
tence. Eur. J. Clin. Nutr. 56 (Suppl 3 ), S42–S45.
McNelis, J.C., and Olefsky, J.M. (2014). Macrophages, immunity, and meta-
Lazo, M., Hernaez, R., Eberhardt, M.S., Bonekamp, S., Kamel, I., Guallar, E., bolic disease. Immunity 41, 36–48.
Koteish, A., Brancati, F.L., and Clark, J.M. (2013). Prevalence of nonalcoholic
fatty liver disease in the United States: the Third National Health and Nutrition Meerzaman, D.M., Yan, C., Chen, Q.R., Edmonson, M.N., Schaefer, C.F., Clif-
Examination Survey, 1988-1994. Am. J. Epidemiol. 178, 38–45. ford, R.J., Dunn, B.K., Dong, L., Finney, R.P., Cultraro, C.M., et al. (2014).
Genome-wide transcriptional sequencing identifies novel mutations in meta-
Lee, J.H., Budanov, A.V., and Karin, M. (2013). Sestrins orchestrate cellular bolic genes in human hepatocellular carcinoma. Cancer Genomics Prote-
metabolism to attenuate aging. Cell Metab. 18, 792–801. omics 11, 1–12.

Li, N., Wu, X., Holzer, R.G., Lee, J.H., Todoric, J., Park, E.J., Ogata, H., Gukov- Menon, S., Yecies, J.L., Zhang, H.H., Howell, J.J., Nicholatos, J., Harputlugil,
skaya, A.S., Gukovsky, I., Pizzo, D.P., et al. (2013). Loss of acinar cell IKKa trig- E., Bronson, R.T., Kwiatkowski, D.J., and Manning, B.D. (2012). Chronic acti-
gers spontaneous pancreatitis in mice. J. Clin. Invest. 123, 2231–2243. vation of mTOR complex 1 is sufficient to cause hepatocellular carcinoma in
mice. Sci. Signal. 5, ra24.
Lim, C., and Savan, R. (2014). The role of the IL-22/IL-22R1 axis in cancer.
Cytokine Growth Factor Rev. 25, 257–271. Moon, H.S., Liu, X., Nagel, J.M., Chamberland, J.P., Diakopoulos, K.N., Brin-
koetter, M.T., Hatziapostolou, M., Wu, Y., Robson, S.C., Iliopoulos, D., and
Lin, Y., Yagyu, K., Egawa, N., Ueno, M., Mori, M., Nakao, H., Ishii, H., Naka- Mantzoros, C.S. (2013). Salutary effects of adiponectin on colon cancer: in vivo
mura, K., Wakai, K., Hosono, S., et al. (2011). An overview of genetic polymor- and in vitro studies in mice. Gut 62, 561–570.
phisms and pancreatic cancer risk in molecular epidemiologic studies.
J. Epidemiol. 21, 2–12. Moscat, J., and Diaz-Meco, M.T. (2012). p62: a versatile multitasker takes on
cancer. Trends Biochem. Sci. 37, 230–236.
Ling, J., Kang, Y., Zhao, R., Xia, Q., Lee, D.F., Chang, Z., Li, J., Peng, B.,
Fleming, J.B., Wang, H., et al. (2012). KrasG12D-induced IKK2/b/NF-kB acti- Mutoh, M., Teraoka, N., Takasu, S., Takahashi, M., Onuma, K., Yamamoto, M.,
vation by IL-1a and p62 feedforward loops is required for development of Kubota, N., Iseki, T., Kadowaki, T., Sugimura, T., and Wakabayashi, K. (2011).
pancreatic ductal adenocarcinoma. Cancer Cell 21, 105–120. Loss of adiponectin promotes intestinal carcinogenesis in Min and wild-type
mice. Gastroenterology 140, 2000–2008, 2008.e1–2008.e2.
Liu, Z., Brooks, R.S., Ciappio, E.D., Kim, S.J., Crott, J.W., Bennett, G., Green-
berg, A.S., and Mason, J.B. (2012). Diet-induced obesity elevates colonic TNF- Nakagawa, H., Umemura, A., Taniguchi, K., Font-Burgada, J., Dhar, D., Ogata,
a in mice and is accompanied by an activation of Wnt signaling: a mechanism H., Zhong, Z., Valasek, M.A., Seki, E., Hidalgo, J., et al. (2014). ER stress co-
for obesity-associated colorectal cancer. J. Nutr. Biochem. 23, 1207–1213. operates with hypernutrition to trigger TNF-dependent spontaneous HCC
development. Cancer Cell 26, 331–343.
Liu, Y.L., Patman, G.L., Leathart, J.B., Piguet, A.C., Burt, A.D., Dufour, J.F.,
Day, C.P., Daly, A.K., Reeves, H.L., and Anstee, Q.M. (2014). Carriage of the Naugler, W.E., Sakurai, T., Kim, S., Maeda, S., Kim, K., Elsharkawy, A.M., and
PNPLA3 rs738409 C >G polymorphism confers an increased risk of non-alco- Karin, M. (2007). Gender disparity in liver cancer due to sex differences in
holic fatty liver disease associated hepatocellular carcinoma. J. Hepatol. 61, MyD88-dependent IL-6 production. Science 317, 121–124.
75–81.
Ng, M., Fleming, T., Robinson, M., Thomson, B., Graetz, N., Margono, C., Mul-
Loos, R.J., Lindgren, C.M., Li, S., Wheeler, E., Zhao, J.H., Prokopenko, I., In- lany, E.C., Biryukov, S., Abbafati, C., Abera, S.F., et al. (2014). Global, regional,
ouye, M., Freathy, R.M., Attwood, A.P., Beckmann, J.S., et al.; Prostate, Lung, and national prevalence of overweight and obesity in children and adults dur-
Colorectal, and Ovarian (PLCO) Cancer Screening Trial; KORA; Nurses’ Health ing 1980-2013: a systematic analysis for the Global Burden of Disease Study
Study; Diabetes Genetics Initiative; SardiNIA Study; Wellcome Trust Case 2013. Lancet 384, 766–781.

60 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.


Cell Metabolism

Review
Nielsen, S., Guo, Z., Johnson, C.M., Hensrud, D.D., and Jensen, M.D. (2004). Rothwell, P.M., Fowkes, F.G., Belch, J.F., Ogawa, H., Warlow, C.P., and
Splanchnic lipolysis in human obesity. J. Clin. Invest. 113, 1582–1588. Meade, T.W. (2011). Effect of daily aspirin on long-term risk of death due to
cancer: analysis of individual patient data from randomised trials. Lancet
Ogden, C.L., Carroll, M.D., Kit, B.K., and Flegal, K.M. (2014). Prevalence of 377, 31–41.
childhood and adult obesity in the United States, 2011-2012. JAMA 311,
806–814. Saitoh, T., Fujita, N., Jang, M.H., Uematsu, S., Yang, B.G., Satoh, T., Omori, H.,
Noda, T., Yamamoto, N., Komatsu, M., et al. (2008). Loss of the autophagy
Otake, S., Takeda, H., Fujishima, S., Fukui, T., Orii, T., Sato, T., Sasaki, Y., protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature
Nishise, S., and Kawata, S. (2010). Decreased levels of plasma adiponectin 456, 264–268.
associated with increased risk of colorectal cancer. World J. Gastroenterol.
16, 1252–1257. Schneider, J.L., and Cuervo, A.M. (2014). Liver autophagy: much more than
just taking out the trash. Nat. Rev. Gastroenterol. Hepatol. 11, 187–200.
Ozcan, U., Yilmaz, E., Ozcan, L., Furuhashi, M., Vaillancourt, E., Smith, R.O.,
Görgün, C.Z., and Hotamisligil, G.S. (2006). Chemical chaperones reduce Schulz, M.D., Atay, C., Heringer, J., Romrig, F.K., Schwitalla, S., Aydin, B., Zie-
ER stress and restore glucose homeostasis in a mouse model of type 2 dia- gler, P.K., Varga, J., Reindl, W., Pommerenke, C., et al. (2014). High-fat-diet-
betes. Science 313, 1137–1140. mediated dysbiosis promotes intestinal carcinogenesis independently of
obesity. Nature 514, 508–512.
Park, E.J., Lee, J.H., Yu, G.Y., He, G., Ali, S.R., Holzer, R.G., Osterreicher,
C.H., Takahashi, H., and Karin, M. (2010). Dietary and genetic obesity promote Schwabe, R.F., and Jobin, C. (2013). The microbiome and cancer. Nat. Rev.
liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cancer 13, 800–812.
Cell 140, 197–208.
Schwimmer, J.B., Celedon, M.A., Lavine, J.E., Salem, R., Campbell, N.,
Pei, Y.F., Zhang, L., Liu, Y., Li, J., Shen, H., Liu, Y.Z., Tian, Q., He, H., Wu, S., Schork, N.J., Shiehmorteza, M., Yokoo, T., Chavez, A., Middleton, M.S., and
Ran, S., et al. (2014). Meta-analysis of genome-wide association data iden- Sirlin, C.B. (2009). Heritability of nonalcoholic fatty liver disease. Gastroenter-
tifies novel susceptibility loci for obesity. Hum. Mol. Genet. 23, 820–830. ology 136, 1585–1592.
Perera, R.M., Stoykova, S., Nicolay, B.N., Ross, K.N., Fitamant, J., Boukhali, Seki, E., De Minicis, S., Gwak, G.Y., Kluwe, J., Inokuchi, S., Bursill, C.A., Llo-
M., Lengrand, J., Deshpande, V., Selig, M.K., Ferrone, C.R., et al. (2015). Tran- vet, J.M., Brenner, D.A., and Schwabe, R.F. (2009). CCR1 and CCR5 promote
scriptional control of autophagy-lysosome function drives pancreatic cancer hepatic fibrosis in mice. J. Clin. Invest. 119, 1858–1870.
metabolism. Nature 524, 361–365.
Sheridan, P.A., Paich, H.A., Handy, J., Karlsson, E.A., Hudgens, M.G., Sam-
Petersen, G.M., Amundadottir, L., Fuchs, C.S., Kraft, P., Stolzenberg-Solo- mon, A.B., Holland, L.A., Weir, S., Noah, T.L., and Beck, M.A. (2012). Obesity
mon, R.Z., Jacobs, K.B., Arslan, A.A., Bueno-de-Mesquita, H.B., Gallinger, is associated with impaired immune response to influenza vaccination in hu-
S., Gross, M., et al. (2010a). A genome-wide association study identifies mans. Int. J. Obes. 36, 1072–1077.
pancreatic cancer susceptibility loci on chromosomes 13q22.1, 1q32.1 and
5p15.33. Nat. Genet. 42, 224–228. Shi, H., Kokoeva, M.V., Inouye, K., Tzameli, I., Yin, H., and Flier, J.S. (2006).
TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin.
Petersen, K.F., Dufour, S., Hariri, A., Nelson-Williams, C., Foo, J.N., Zhang, Invest. 116, 3015–3025.
X.M., Dziura, J., Lifton, R.P., and Shulman, G.I. (2010b). Apolipoprotein C3
gene variants in nonalcoholic fatty liver disease. N. Engl. J. Med. 362, 1082– Solinas, G., Naugler, W., Galimi, F., Lee, M.S., and Karin, M. (2006). Saturated
1089. fatty acids inhibit induction of insulin gene transcription by JNK-mediated
phosphorylation of insulin-receptor substrates. Proc. Natl. Acad. Sci. USA
Poullis, A., Foster, R., Shetty, A., Fagerhol, M.K., and Mendall, M.A. (2004).
103, 16454–16459.
Bowel inflammation as measured by fecal calprotectin: a link between lifestyle
factors and colorectal cancer risk. Cancer Epidemiol. Biomarkers Prev. 13,
Solinas, G., Vilcu, C., Neels, J.G., Bandyopadhyay, G.K., Luo, J.L., Naugler,
279–284.
W., Grivennikov, S., Wynshaw-Boris, A., Scadeng, M., Olefsky, J.M., and
Karin, M. (2007). JNK1 in hematopoietically derived cells contributes to diet-
Qu, X., Zou, Z., Sun, Q., Luby-Phelps, K., Cheng, P., Hogan, R.N., Gilpin, C.,
induced inflammation and insulin resistance without affecting obesity. Cell
and Levine, B. (2007). Autophagy gene-dependent clearance of apoptotic cells
Metab. 6, 386–397.
during embryonic development. Cell 128, 931–946.

Quinn, B.J., Kitagawa, H., Memmott, R.M., Gills, J.J., and Dennis, P.A. (2013). Solomon, D.H., Massarotti, E., Garg, R., Liu, J., Canning, C., and Schnee-
Repositioning metformin for cancer prevention and treatment. Trends Endocri- weiss, S. (2011). Association between disease-modifying antirheumatic drugs
nol. Metab. 24, 469–480. and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA
305, 2525–2531.
Rahib, L., Smith, B.D., Aizenberg, R., Rosenzweig, A.B., Fleshman, J.M., and
Matrisian, L.M. (2014). Projecting cancer incidence and deaths to 2030: the un- Sookoian, S., and Pirola, C.J. (2011). Meta-analysis of the influence of I148M
expected burden of thyroid, liver, and pancreas cancers in the United States. variant of patatin-like phospholipase domain containing 3 gene (PNPLA3) on
Cancer Res. 74, 2913–2921. the susceptibility and histological severity of nonalcoholic fatty liver disease.
Hepatology 53, 1883–1894.
Reinehr, T., Roth, C., Menke, T., and Andler, W. (2004). Adiponectin before and
after weight loss in obese children. J. Clin. Endocrinol. Metab. 89, 3790–3794. Speliotes, E.K., Yerges-Armstrong, L.M., Wu, J., Hernaez, R., Kim, L.J.,
Palmer, C.D., Gudnason, V., Eiriksdottir, G., Garcia, M.E., Launer, L.J., et al.;
Rhim, A.D., Mirek, E.T., Aiello, N.M., Maitra, A., Bailey, J.M., McAllister, F., NASH CRN; GIANT Consortium; MAGIC Investigators; GOLD Consortium
Reichert, M., Beatty, G.L., Rustgi, A.K., Vonderheide, R.H., et al. (2012). (2011). Genome-wide association analysis identifies variants associated with
EMT and dissemination precede pancreatic tumor formation. Cell 148, nonalcoholic fatty liver disease that have distinct effects on metabolic traits.
349–361. PLoS Genet. 7, e1001324.

Rocchini, A.P. (2002). Childhood obesity and a diabetes epidemic. N. Engl. J. Stanley, T.L., Zanni, M.V., Johnsen, S., Rasheed, S., Makimura, H., Lee, H.,
Med. 346, 854–855. Khor, V.K., Ahima, R.S., and Grinspoon, S.K. (2011). TNF-alpha antagonism
with etanercept decreases glucose and increases the proportion of high mo-
Roden, M., Price, T.B., Perseghin, G., Petersen, K.F., Rothman, D.L., Cline, lecular weight adiponectin in obese subjects with features of the metabolic
G.W., and Shulman, G.I. (1996). Mechanism of free fatty acid-induced insulin syndrome. J. Clin. Endocrinol. Metab. 96, E146–E150.
resistance in humans. J. Clin. Invest. 97, 2859–2865.
Sugiyama, M., Takahashi, H., Hosono, K., Endo, H., Kato, S., Yoneda, K., No-
Romeo, S., Kozlitina, J., Xing, C., Pertsemlidis, A., Cox, D., Pennacchio, L.A., zaki, Y., Fujita, K., Yoneda, M., Wada, K., et al. (2009). Adiponectin inhibits
Boerwinkle, E., Cohen, J.C., and Hobbs, H.H. (2008). Genetic variation in colorectal cancer cell growth through the AMPK/mTOR pathway. Int. J. Oncol.
PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 34, 339–344.
40, 1461–1465.
Takamura, A., Komatsu, M., Hara, T., Sakamoto, A., Kishi, C., Waguri, S., Eishi,
Rosen, E.D., and Spiegelman, B.M. (2014). What we talk about when we talk Y., Hino, O., Tanaka, K., and Mizushima, N. (2011). Autophagy-deficient mice
about fat. Cell 156, 20–44. develop multiple liver tumors. Genes Dev. 25, 795–800.

Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc. 61


Cell Metabolism

Review
Tanaka, S., Isoda, F., Kiuchi, Y., Ikeda, H., Mobbs, C.V., and Yamakawa, T. White, E., Karp, C., Strohecker, A.M., Guo, Y., and Mathew, R. (2010). Role of
(2000). T lymphopenia in genetically obese-diabetic Wistar fatty rats: effects autophagy in suppression of inflammation and cancer. Curr. Opin. Cell Biol.
of body weight reduction on T cells. Metabolism 49, 1261–1266. 22, 212–217.

Tang, H., Dong, X., Hassan, M., Abbruzzese, J.L., and Li, D. (2011). Body mass WHO Expert Consultation (2004). Appropriate body-mass index for Asian pop-
index and obesity- and diabetes-associated genotypes and risk for pancreatic ulations and its implications for policy and intervention strategies. Lancet 363,
cancer. Cancer Epidemiol. Biomarkers Prev. 20, 779–792. 157–163.

Taniguchi, K., and Karin, M. (2014). IL-6 and related cytokines as the critical Willer, C.J., Speliotes, E.K., Loos, R.J., Li, S., Lindgren, C.M., Heid, I.M.,
lynchpins between inflammation and cancer. Semin. Immunol. 26, 54–74. Berndt, S.I., Elliott, A.L., Jackson, A.U., Lamina, C., et al.; Wellcome Trust
Case Control Consortium; Genetic Investigation of ANthropometric Traits
Teixeira, T.F., Collado, M.C., Ferreira, C.L., Bressan, J., and Peluzio, Mdo.C. Consortium (2009). Six new loci associated with body mass index highlight a
(2012). Potential mechanisms for the emerging link between obesity and neuronal influence on body weight regulation. Nat. Genet. 41, 25–34.
increased intestinal permeability. Nutr. Res. 32, 637–647.
Wiseman, M. (2008). The second World Cancer Research Fund/American
Tilg, H., and Kaser, A. (2011). Gut microbiome, obesity, and metabolic Institute for Cancer Research expert report. Food, nutrition, physical activity,
dysfunction. J. Clin. Invest. 121, 2126–2132. and the prevention of cancer: a global perspective. Proc. Nutr. Soc. 67,
253–256.
Tremaroli, V., and Bäckhed, F. (2012). Functional interactions between the gut
microbiota and host metabolism. Nature 489, 242–249. Wolf, M.J., Adili, A., Piotrowitz, K., Abdullah, Z., Boege, Y., Stemmer, K., Ring-
elhan, M., Simonavicius, N., Egger, M., Wohlleber, D., et al. (2014). Metabolic
Tremaroli, V., Karlsson, F., Werling, M., Ståhlman, M., Kovatcheva-Datchary, activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic
P., Olbers, T., Fändriks, L., le Roux, C.W., Nielsen, J., and Bäckhed, F. steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell
(2015). Roux-en-Y Gastric Bypass and Vertical Banded Gastroplasty Induce 26, 549–564.
Long-Term Changes on the Human Gut Microbiome Contributing to Fat
Mass Regulation. Cell Metab. 22, 228–238. Wu, C., Miao, X., Huang, L., Che, X., Jiang, G., Yu, D., Yang, X., Cao, G., Hu, Z.,
Zhou, Y., et al. (2012). Genome-wide association study identifies five loci asso-
Turnbaugh, P.J., Ley, R.E., Mahowald, M.A., Magrini, V., Mardis, E.R., and ciated with susceptibility to pancreatic cancer in Chinese populations. Nat.
Gordon, J.I. (2006). An obesity-associated gut microbiome with increased ca- Genet. 44, 62–66.
pacity for energy harvest. Nature 444, 1027–1031.
Xu, H., Barnes, G.T., Yang, Q., Tan, G., Yang, D., Chou, C.J., Sole, J., Nichols,
Uddin, S., Hussain, A.R., Siraj, A.K., Khan, O.S., Bavi, P.P., and Al-Kuraya, K.S. A., Ross, J.S., Tartaglia, L.A., and Chen, H. (2003). Chronic inflammation in fat
(2011). Role of leptin and its receptors in the pathogenesis of thyroid cancer. plays a crucial role in the development of obesity-related insulin resistance.
Int. J. Clin. Exp. Pathol. 4, 637–643. J. Clin. Invest. 112, 1821–1830.

Umemura, A., Park, E.J., Taniguchi, K., Lee, J.H., Shalapour, S., Valasek, M.A., Xu, X., Grijalva, A., Skowronski, A., van Eijk, M., Serlie, M.J., and Ferrante,
Aghajan, M., Nakagawa, H., Seki, E., Hall, M.N., and Karin, M. (2014). Liver A.W., Jr. (2013). Obesity activates a program of lysosomal-dependent lipid
damage, inflammation, and enhanced tumorigenesis after persistent mTORC1 metabolism in adipose tissue macrophages independently of classic activa-
inhibition. Cell Metab. 20, 133–144. tion. Cell Metab. 18, 816–830.

Upadhyay, V., Poroyko, V., Kim, T.J., Devkota, S., Fu, S., Liu, D., Tumanov, Yamauchi, T., Kamon, J., Minokoshi, Y., Ito, Y., Waki, H., Uchida, S., Yama-
A.V., Koroleva, E.P., Deng, L., Nagler, C., et al. (2012). Lymphotoxin regulates shita, S., Noda, M., Kita, S., Ueki, K., et al. (2002). Adiponectin stimulates
commensal responses to enable diet-induced obesity. Nat. Immunol. 13, glucose utilization and fatty-acid oxidation by activating AMP-activated pro-
947–953. tein kinase. Nat. Med. 8, 1288–1295.

Uusitupa, M., Lindi, V., Louheranta, A., Salopuro, T., Lindström, J., and Tuomi- Yan, B., Wang, H., Rabbani, Z.N., Zhao, Y., Li, W., Yuan, Y., Li, F., Dewhirst,
lehto, J.; Finnish Diabetes Prevention Study Group (2003). Long-term improve- M.W., and Li, C.Y. (2006). Tumor necrosis factor-alpha is a potent endogenous
ment in insulin sensitivity by changing lifestyles of people with impaired mutagen that promotes cellular transformation. Cancer Res. 66, 11565–11570.
glucose tolerance: 4-year results from the Finnish Diabetes Prevention Study.
Diabetes 52, 2532–2538. Yang, Y., Ju, D., Zhang, M., and Yang, G. (2008). Interleukin-6 stimulates lipol-
ysis in porcine adipocytes. Endocrine 33, 261–269.
Uysal, K.T., Wiesbrock, S.M., Marino, M.W., and Hotamisligil, G.S. (1997). Pro-
tection from obesity-induced insulin resistance in mice lacking TNF-alpha Yang, L., Li, P., Fu, S., Calay, E.S., and Hotamisligil, G.S. (2010). Defective he-
function. Nature 389, 610–614. patic autophagy in obesity promotes ER stress and causes insulin resistance.
Cell Metab. 11, 467–478.
Vincent, A., Herman, J., Schulick, R., Hruban, R.H., and Goggins, M. (2011).
Pancreatic cancer. Lancet 378, 607–620. Yang, L., Calay, E.S., Fan, J., Arduini, A., Kunz, R.C., Gygi, S.P., Yalcin, A., Fu,
S., and Hotamisligil, G.S. (2015). METABOLISM. S-Nitrosylation links obesity-
Wang, X., Ota, N., Manzanillo, P., Kates, L., Zavala-Solorio, J., Eidenschenk, associated inflammation to endoplasmic reticulum dysfunction. Science 349,
C., Zhang, J., Lesch, J., Lee, W.P., Ross, J., et al. (2014). Interleukin-22 allevi- 500–506.
ates metabolic disorders and restores mucosal immunity in diabetes. Nature
514, 237–241. Yen, W.L., and Klionsky, D.J. (2008). How to live long and prosper: autophagy,
mitochondria, and aging. Physiology (Bethesda) 23, 248–262.
Warburg, O. (1956). On the origin of cancer cells. Science 123, 309–314.
Yoshimoto, S., Loo, T.M., Atarashi, K., Kanda, H., Sato, S., Oyadomari, S.,
Waris, G., Tardif, K.D., and Siddiqui, A. (2002). Endoplasmic reticulum (ER) Iwakura, Y., Oshima, K., Morita, H., Hattori, M., et al. (2013). Obesity-induced
stress: hepatitis C virus induces an ER-nucleus signal transduction pathway gut microbial metabolite promotes liver cancer through senescence secre-
and activates NF-kappaB and STAT-3. Biochem. Pharmacol. 64, 1425–1430. tome. Nature 499, 97–101.

Watanabe, R.M. (2010). The genetics of insulin resistance: Where’s Waldo? Zatloukal, K., Stumptner, C., Fuchsbichler, A., Heid, H., Schnoelzer, M., Ken-
Curr. Diab. Rep. 10, 476–484. ner, L., Kleinert, R., Prinz, M., Aguzzi, A., and Denk, H. (2002). p62 Is a common
component of cytoplasmic inclusions in protein aggregation diseases. Am. J.
Weisberg, S.P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R.L., and Fer- Pathol. 160, 255–263.
rante, A.W., Jr. (2003). Obesity is associated with macrophage accumulation in
adipose tissue. J. Clin. Invest. 112, 1796–1808. Zhang, H.H., Halbleib, M., Ahmad, F., Manganiello, V.C., and Greenberg, A.S.
(2002). Tumor necrosis factor-alpha stimulates lipolysis in differentiated hu-
Wen, W., Cho, Y.S., Zheng, W., Dorajoo, R., Kato, N., Qi, L., Chen, C.H., De- man adipocytes through activation of extracellular signal-related kinase and
lahanty, R.J., Okada, Y., Tabara, Y., et al.; Genetic Investigation of ANthropo- elevation of intracellular cAMP. Diabetes 51, 2929–2935.
metric Traits (GIANT) Consortium (2012). Meta-analysis identifies common
variants associated with body mass index in east Asians. Nat. Genet. 44, Zhou, R., Yazdi, A.S., Menu, P., and Tschopp, J. (2011). A role for mitochondria
307–311. in NLRP3 inflammasome activation. Nature 469, 221–225.

62 Cell Metabolism 23, January 12, 2016 ª2016 Elsevier Inc.

You might also like