Sie sind auf Seite 1von 16

Rev Endocr Metab Disord (2016) 17:389–403

DOI 10.1007/s11154-016-9393-9

Obesity as a risk factor for malignant melanoma


and non-melanoma skin cancer
K. Karimi 1 & T. H. Lindgren 1 & C. A. Koch 2,3,4 & Robert T. Brodell 5,6,7

Published online: 10 November 2016


# Springer Science+Business Media New York 2016

Abstract The dramatic increases in incidence of both obesity 1 Introduction


and many cancers including skin cancer emphasize the need to
better understand the pathophysiology of both conditions and Obesity is a leading cause of chronic illness and morbidity.
their connections. Melanoma is considered the fastest growing It contributes heavily to both individual and societal health
cancer and rates of non-melanoma skin cancer have also in- expenditures, and accounts for nearly 20 % of all cancer cases
creased over the last decade. The molecular mechanisms un- [1]. A variety of factors are at play in the progression of cancer
derlying the association between obesity and skin cancer are in patients with obesity and/or type 2 diabetes [2]. Increased
not clearly understood but emerging evidence points to chang- weight or height directly correlates with the number of total
es in the tumor microenvironment including aberrant cell sig- cells available for malignant transformation [3]. In addition,
naling and genomic instability in the chronic inflammatory cancer may be caused by obesity-induced modifications of the
state many obese individuals experience. This article reviews immune system or long-term storage of toxins or medications
the literature linking obesity to melanoma and non-melanoma in adipose tissue [4, 5]. Whether obesity increases the risk of
skin cancer. melanoma or non-melanoma skin cancers (NMSC) is less well
established.
Several studies have demonstrated a link between malig-
Keywords Melanoma . Obesity . Non-melanoma skin nant melanoma and excess adiposity [6–11], although a large
cancer . Basal cell carcinoma . Squamous cell carcinoma . study demonstrated no association [12, 13]. Other studies sug-
Inflammation . Leptin . Insulin gested obesity is associated with an increased risk of malig-
nant melanoma in males [14–16] but not females [13, 14,
* Robert T. Brodell 16–20]. Similarly, there is conflicting evidence with regard
rbrodell@umc.edu to obesity and NMSC. In one study, excess body weight and
body mass index (BMI) were inversely associated with the
1
School of Medicine, Texas Tech University Health Sciences Center, development of NMSC [12, 13], while a positive association
Lubbock, TX, USA between obesity and NMSC has also been described [21, 22].
2
Division of Endocrinology, University of Mississippi Medical Courneya and colleagues proposed that obesity may be a sig-
Center, Jackson, MS, USA nificant risk factor for the development of skin cancer only in
3
Cancer Institute, University of Mississippi Medical Center, areas with lower UV radiation. The impact of higher UV ra-
Jackson, MS, USA diation inducing NMSC may overshadow the less significant
4
G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS, USA effects of obesity [21].
5
Department of Dermatology, University of Mississippi Medical
Center, 2500 North State Street, Jackson, MS 39216, USA
6
Department of Pathology, University of Mississippi Medical Center, 2 Obesity
Jackson, MS, USA
7
Department of Dermatology, University of Rochester School of Obesity rates have dramatically increased in the USA over the
Medicine and Dentistry, Rochester, NY, USA past three decades, doubling in the last 25 years [23, 24].
390 Rev Endocr Metab Disord (2016) 17:389–403

According to the US Centers for Disease Control and 2.2 Obesity and oxidative stress
Prevention (CDC), 35 % of the US population is obese, and
6 % of the population is extremely obese (Obesity Prevalence Systemic oxidative stress is another characteristic of obesity
Maps, Centers for Disease Control and Prevention [46]. Obese subjects exhibit reductions in antioxidant capaci-
http://www.cdc.gov/obesity/data/prevalence-maps.html). ty, elevated reactive oxygen species (ROS) production, and
The 13th annual report from Trust for America’s Health increased serum lipid oxidation products [47–49]. Adipose
and the Robert Wood Johnson Foundation found that rates tissue itself appears to be the major source of systemic oxida-
of obesity now exceed 35 % in four states, are at or above tive stress in obesity [50]. Additionally, as expansion of adi-
30 % in 25 states, and are above 20 % in all states pocytes occurs, the distance between adipocytes and capil-
(http://healthyamericans.org/reports/stateofobesity2016). laries increases, leading to adipose tissue hypoxia [46]. In
Even more troubling, the prevalence of obesity has tripled in mice, adipose tissue hypoxia has been shown to further pro-
children and adolescents over the past 40 years. Obesity mote systemic oxidative stress by suppression of glutathione
prevalence is also increasing in both developed and peroxidase 3 (GPx3), leading to exacerbation of glucose-
developing nations [25, 26]. induced insulin resistance [46].
The cause of obesity is not clear, but interactions between
environmental factors (e.g., gut microbiota, ambient tempera-
ture, energy balance, excessive caloric intake, and insufficient 2.2.1 Diet and obesity
physical activity) and genetic factors, some reflected by pig-
mentation [27–31], play a role. The genes that regulate tan- High-fat, high-carbohydrate, and high-caloric diets contribute
ning ability and pigmentation of hair, skin, and eyes may be to obesity. Cancer can also be influenced by diet. Excessive
associated with intrinsic factors that produce obesity [32, 33]. meat consumption (i.e., greater than 5 servings per day), in
Infections may also contribute to obesity [34]. Adipocytes and particular, is associated with greater incidence of cancer be-
macrophages are similar [35], and pre-adipocytes have the cause of environmental carcinogens present in meat and char-
ability to rapidly and efficiently differentiate into macro- cuterie products [51]. Specifically, cooked fish and beef con-
phages, thereby linking fat and innate immune processes tain heterocyclic amines called aminoimidazole-arenes
[35]. Furthermore, adipose tissue may expand in response to (AIAs) that are pro-mutagens and pro-carcinogens [52].
infection [34]. While these factors are present in low levels in these foods,
excessive consumption may induce carcinogenic effects in
obese individuals.
2.1 Obesity and inflammation

Inflammation may be associated with obesity, reflecting 2.2.2 Genetics and obesity
the body’s response to pro-inflammatory cytokines [34,
36]. Pro-inflammatory markers like macrophage colony- Ethnicity and specific gene pathways contribute to the
stimulating factor (MCSF) can predict obesity in human development of obesity. Several causal single nucleotide
subjects [37, 38]. In both diet-induced and genetic obesity polymorphisms (SNPs), including those in or near the
[39], adipose tissue becomes infiltrated with adipose tis- mitochondrial carrier 2 (MTCH2) and melanocortin 4
sue macrophages (ATM). These ATMs produce pro- receptor (MC4R) genes, are associated with obesity.
inflammatory cytokines including interleukin 8 (IL-8), These SNPs are more significant in Caucasian and
monocyte chemo-attractant protein-1 (MCP-1), tumor ne- Hispanic subjects compared to Chinese and African
crosis factor alpha (TNF-alpha), interleukin 6 (IL-6), and American people [53]. Furthermore, total cholesterol,
complement proteins [40–42]. Leukocytes are also in- which is directly related to obesity, is higher in Black
creased [43], reflecting low-grade systemic inflammation. children, who are also significantly more likely to be
Inflammatory kinases further amplify inflammation in obese than White or Asian children [54].
obese individuals [44]. The rapamycin (MTOR) pathway may link obesity and
In addition to promoting obesity-induced angiogenesis, in- cancer mechanisms. As a regulator of cell growth, prolifera-
flammation may also promote tumorigenesis and melanoma tion, and survival, the MTOR gene is activated by oxidative
growth [45]. Adipocytes secrete inflammatory cytokines that stress, which is increased in obese individuals [55]. Under
recruit macrophages to initiate angiogenesis [24]. stress, the mTOR gene promotes cell survival and proliferation
Macrophages may play a central role in both angiogenesis of as well as gene transcription to metabolize carbohydrates, in-
melanoma and of obesity via upregulation of the vascular crease lipogenesis, and inhibit autophagy [55]. Thus, MTOR
endothelial growth factor (VEGF) pathway accelerated by allows for the proliferation and survival of cancer cells and
leptin [24]. promotes metastases.
Rev Endocr Metab Disord (2016) 17:389–403 391

2.3 Obesity and the immune system 2.5 Obesity and insulin

Adipose tissue inflammation is a risk factor for developing and Inflammatory cytokines from adipose tissue produce chronic
advancing breast cancer because of adipose tissue’s tendency to inflammation that can induce insulin resistance and type 2
sequester the proangiogenic factor MCP-1 [56]. MCP-1 is a diabetes mellitus [74, 75]. This results in increased pancreatic
chemokine that regulates the migration of monocytes and mac- insulin secretion to compensate for the cells’ lack of insulin
rophages and is involved in cancer pathology. Diet-induced uptake. Hyperinsulinemia then can induce tumorigenesis [2].
obese mice were also found to have variable carcinogenesis- Cancer cells react with highly concentrated insulin in the
related gene expression: cellular proliferation, antioxidant and ex trace llular-sign al-re gula te d k inase (ERK) a nd
DNA protection, and tumor suppression genes [57]. phosphatidylinositol-3 kinase (PI-3K) pathways [76].
ROS are important in cellular communication and homeo- Malignant cancer cells also overexpress insulin receptors, pro-
stasis; however, an inappropriate increase in ROS in the body moting insulin-induced cell proliferation [76].
can contribute to chronic diseases like type 2 diabetes and Hyperinsulinemia in obese individuals and type 2 diabetics
cancer [58]. Oxidative stress can induce carcinogenesis as has been associated with acanthosis nigricans, acrochordons,
well as reduce the effects of anti-oxidants [59, 60]. In an an- finger pebbles, and Leser-Trelat, highlighting the link between
imal study, researchers discovered that obese mice were more insulin level and cell proliferation [77].
likely to exhibit immune system deterioration [61]. This pre- Adiponectin, an adipokine that opposes insulin resistance,
mature immunosenescence suggests a link between obesity is significantly decreased in obese individuals, increasing can-
and an impaired immune system. Interestingly, inflammation cer risk [76].
responses in the body also produce the same exogenous oxi- Insulin-like growth factor-1 is elevated in patients with
dative stress in melanocytes that elicits generation of ROS in acromegaly which is caused by growth hormone excess and
melanocytes [62]. Therefore, a link between obesity- can lead to secondary diabetes mellitus [78]. Acromegalics
associated inflammation and ROS accumulation contributing with diabetes mellitus are three times more likely to develop
to melanoma pathogenesis can be suggested. malignant tumors [79].
The obese mutation in the leptin gene can also contribute to
obesity. Leptin, a hormone that regulates appetite, is reduced 2.6 Obesity and hypoxemia
in people with obesity [44, 63] and is positively associated
with melanoma risk [64]. Leptin mediates macrophage re- Hypoxemia is a commonly underappreciated feature of obe-
sponse to ROS intracellularly [65] and induces oxidative sity that results from over perfusion of poorly ventilated lung
stress via TNF-alpha [66]. Decreased leptin or leptin resis- regions [46]. In many obese patients, closing volume exceeds
tance results in an impaired ability to maintain immunity in functional residual capacity (FRC), and tidal breath is initiated
people with obesity. Interestingly, leptin is also a reliable tu- from volumes at which many airways in the inferior parts of
mor marker of malignant melanoma, as its expression is sig- the lung are closed [46]. Hypoxia enhances systemic inflam-
nificantly increased in human melanoma cells [67]. The inter- mation, impacting all organ systems [46]. Chronic hypox-
play between these factors suggests obesity as an important emia, in turn, increases plasma leptin levels [80].
risk factor for melanoma.

2.4 Obesity and vitamin D 3 Melanoma

Vitamin D has a positive, pro-inflammatory role in white ad- Melanoma is the most aggressive form of skin cancer (Fig. 1),
ipose tissue [68], and obese people often have an accompany- and it most commonly develops from malignant transforma-
ing vitamin D deficiency [69]. This deficiency may be respon- tion of melanocytes at the epidermal-dermal junction [81].
sible for 20 % of obesity-related cancer risk [70]. However, The incidence of malignant melanoma has been increasing
the causal relationship between vitamin D deficiency and obe- consistently since the 1950s [82–85]. Over a 10-year period,
sity has yet to be determined (i.e., whether vitamin D is a melanoma incidence rates doubled in all socioeconomic
consequence of obesity or a risk factor for increased adiposi- groups [86]. While patients with a higher SES are more likely
ty). People with obesity may consume fewer foods that con- to be diagnosed with melanoma, individuals with a lower SES
tain vitamin D or avoid exposed to the sun due to a sedentary or those without health insurance are more likely to have ad-
lifestyle. Vitamin D may also lower leptin concentrations, vanced stage melanoma at diagnosis and are more likely to die
impacting appetite [69]. Finally, adipose tissue traps vitamin from melanoma [86–89]. While sun exposure behaviors are a
D, lowering serum vitamin D concentrations [71]. Both vita- major factor affecting the melanoma epidemic [90], part of the
min D deficiencies [72] and obesity [73] have been associated recorded increase may be the result of expansions in screen-
with a higher risk of cancer. ing, biopsy, and reporting to cancer registries [91–94].
392 Rev Endocr Metab Disord (2016) 17:389–403

Table 2 Other possible risk factors for the development of malignant


melanoma

Cosmetics [110]
Lightening agent:
Rhododendrol [110]
Preservatives [111]:
Parabens [112]
Estrogenic molecules [113]
Environmental exposures
Heavy metals (Cu, Fe, Mn, and Co) [114–118]
Non-metals in pesticides, herbicides, dyes, organic
amines, and polychlorinated phenyls [114–118]
Obesity and increased serum leptin levels [64]

pigmentation; this suggests several possible links to obe-


sity [119]. In fact, a genetic link between obesity and
Fig. 1 This 3 × 3 cm black lesion shows asymmetry, irregular borders,
pigmentation [120–124] has been suggested by linking
variegation of color (tan and black) has been enlarging over several years.
A brown, waxy stuck-on seborrheic keratosis is present nearby early-onset obesity with red hair color in patients with
pro-opiomelanocortin (POMC) mutations [119, 125].
3.1 Risk factors Features of POMC gene mutations include severe obesity,
phototype I skin, and red hair [126, 127]. POMC gene
The well-accepted risk factors for melanoma are detailed in transcriptional activation follows DNA damage induced
Table 1. Less well-accepted risk factors including obesity are by UV radiation in epidermal melanocytes [128, 129].
highlighted in Table 2. Furthermore, increased expression of corticotropin-
releasing hormone, which may stimulate POMC expres-
sion, may serve as a melanoma growth factor, as there is
evidence that it is present in pigmented lesions [130]. A
4 Mechanisms for the association of melanoma variety of POMC peptides are made by different cell
and obesity types. Thus, the prohormone gene exhibits a wide range
of control over multiple physiologic functions [119]
4.1 Melanocortin system and the POMC gene (Fig. 2).
MSH and ACTH, post-translational products of the
Melanocortins play a role in energy homeostasis, steroido- POMC prohormone, are produced locally in the skin and
g en es i s, i nf l am mat i on , i m m un om od ula tio n , an d bind to melanocortin 1 receptor (MC1R) on melanocytes
to increase melanin production [119]; a POMC deficiency
Table 1 Well-known risk factors for the development of malignant results in fair skin and red hair [126] and an increased risk
melanoma
for both malignant melanoma and NMSC [126].
Greater than 25 nevi [95] Additionally, MSH binds to feeding centers like
Fair eye color (blue, green, and hazel) [96] melanocortin 4 receptor (MC4R) to decrease feeding
Light hair color (red > blond > light brown) [96] [127, 131]; therefore, POMC deficiency also results in
Family history melanoma [96] obesity. This suggests that melanoma and obesity are not
Personal or family history of non-melanoma skin cancera [97] directly related, but rather that a third instigate, POMC
Host genome [98] deficiency, links their development.
Childhood cancer [99] POMC neurons are the target of circulating leptin, further
Ultraviolet related suggesting that body weight and skin pigmentation are cru-
Intermittent sun exposure and sun burn [100] cially linked [131]. Alterations in the leptin pathway may
Living in lower latitudes [100] correlate with a cutaneous carcinogenic cytokine response to
Living at higher altitude [101–103] UV radiation [132].
Indoor tanning [104]
Cutaneous melanomas can be divided into the subtypes
Alcohol consumption [105–109]
NF1, RAS (N/H/K), BRAF, and Triple-WT (lack of hot spot
mutations in NF1, RAS (N/H/K), BRAF) with immune infiltra-
a
This association could be the result of detection bias [98] tion being associated with improved survival [133].
Rev Endocr Metab Disord (2016) 17:389–403 393

Lepn decreasing body weight [149, 151–154]. Leptin resistance can


result from defects in leptin transport [155–157], impaired
LEPRb signaling [158, 159], neuronal energy imbalance
[149], or endoplasmic reticulum (ER) stress. Leptin deficiency
LEPRb
LEPRb and leptin resistance are risk factors for obesity [149,
160–162] and are associated with insulin resistance [163,
AgRP
POMC neurons 164] and hypogonadism [161, 165, 166].
neurons Leptin activates leptin receptor long isoform (LEPRb) in the
hypothalamus, acting on both POMC neurons and agouti-
related protein (AgRP) neurons [149]. This results in increased
Neuropepde Y
POMC activation of melanocortin 3 (MC3R) and MC4R to suppress
appetite [162, 167] and inhibit feeding behaviors [149, 168].
Appete
Leptin resistance promotes ER stress and chronic inflam-
ACTH α-MSH
mation that contribute to insulin resistance [169–171].
Notably, ER stress inhibits leptin signaling [172, 173].

MC1R MC3R * MC4R * 4.2.2 Role of leptin in melanoma

Increased circulating leptin results in metabolic disturbances,


Melanin Appete that are compounded by inflammation [163, 174]. Leptin is
structurally similar to pro-inflammatory cytokines and may
modulate CRP [174, 175]. It is therefore plausible that the
* Deleon of these receptors results in Lepn Resistance and obesity
positive associations found between serum leptin levels and
Fig. 2 Leptin and POMC and AgRP neuron interactions skin cancer may be due to inflammation secondary to leptin
resistance and obesity (Fig. 4).
4.2 Increased leptin Leptin may also be a cause of melanoma growth via in-
creased nitric oxide (NO) production and increased levels of
Obesity-associated cancers demonstrate increased leptin circulating endothelial progenitor cells (EPCs), ultimately pro-
levels [134–137] because of leptin resistance in many obese moting angiogenesis [176] and vasculogenesis [177] mediated
people. Leptin levels are positively correlated with BMI, total by VEGF and endogenous fibroblast growth factor 2 (FGF-2)
abdominal fat volume, and subcutaneous fat volume [138]. In [178, 179].
fact, one of the major factors influencing plasma leptin con- Leptin acts as a pro-inflammatory adipokine that influences
centrations is adipose tissue mass [134]. Adipocyte synthesis cytokine production, cellular immunity, and inflammation
of leptin is influenced by several factors, including insulin [180]. Additionally, leptin decreases the expression of the tu-
[139], TNF-alpha [140], glucocorticoids [141], gonadal hor- mor suppressor p53 [181] to promote cell cycle progression.
mones [142], catecholamines, and prostaglandins [143]. Some Furthermore, melanoma cells, but not melanocytes, express
of these factors have been associated with neoplastic process- the leptin protein, ultimately creating a positive autocrine
es (Fig. 3). feedback loop [81] that contributes to uncontrolled prolifera-
Leptin also promotes cell division, proliferation, and pos- tion of melanoma tumor cells.
sible melanoma metastasis [64, 144]. High fat diet may further Epidermal hypoxia reduces apoptosis and favors survival
promote melanoma progression [145]. High serum leptin is of melanoma cells [182]. Hypoxic conditions also increase
positively correlated with melanoma risk [64] by accelerating leptin levels via hypoxia-inducible factor 1-alpha (HIF-1-al-
melanoma tumor growth [24]. Furthermore, leptin antagonist pha) [183]. In addition to stimulation by hypoxia, HIF-1-alpha
antibodies have been found to decrease melanoma progres- is also activated by insulin, insulin-like growth factor (IGF),
sion [146]. The correlation between serum leptin levels and angiotensin II, thrombin, platelet-derived growth factor
melanoma risk requires further study to determine if leptin (PDGF), endothelin-1, interleukins, and TNF-alpha [183].
plays a significant role in the development of melanoma.
4.3 Other connections between obesity and melanoma
4.2.1 Role of leptin in obesity
4.3.1 Decreased adiponectin
Leptin inhibits insulin secretion and lipogenesis [147, 148],
stimulates lipolysis and fatty acid oxidation [149, 150], sup- Adiponectin has anti-tumor effects by inhibiting cell prolifer-
presses appetite, and promotes energy expenditure, ultimately ation [184–189]. Adiponectin levels are approximately 50 %
394 Rev Endocr Metab Disord (2016) 17:389–403

Fig. 3 Regulatory pathways of Insulin Glucocorcoids


leptin TNF-α
Gonadal Prostaglandins
hormones Catecholamines

Adipocyte
Lipolysis
Energy B-Oxidaon
Body Obese
Expenditure gene
Weight Pancreac B Insulin
Cells
Vitamin D
Lepn

Serum Macrophages ROS


FGF2 Nitric Oxide
VEGF EPCs
Hypothalamus Cytokines
Angiogenesis &
Vasculogenesis Skin

Appete Inflammaon
Proliferaon
Melanin
Tumor Growth Weight Gain Obesity

Lepn
Anbodies Melanoma

lower in obese individuals [189]; this allows for increased cell may contribute to an increased cancer risk and cancer progres-
proliferation. Mantzoros and colleagues, however, found that sion; on the other hand, calorie restriction, intentional weight
the inverse relationship between serum adiponectin and mel- loss, and diabetes treatments may reduce the risk and rate of
anoma is not significant [190]. skin cancer progression [2, 192]. The IGF-1 receptor, in par-
ticular, inhibits cell proliferation, differentiation, and survival
4.3.2 Insulin resistance and insulin-like growth factor in the skin, and its inactivation results in abnormal differenti-
ation patterns in keratinocytes [193].
Increased insulin, IGF-1, and IGF-2 are sufficient to induce Another pathway involved in insulin resistance is the phos-
tumorigenesis via the insulin receptor [191]. Inflammatory phatidylinositol 3-kinase/protein kinase B (PI3K/Akt) path-
kinases inhibit insulin action and glucose uptake [44] in obese way in which a PI3K inhibitor and an MTOR inhibitor result
individuals. Hyperinsulinemia and increased levels of IGF-1 in the proliferation and prolonged survival of melanoma cells

Genec UV Radiaon Neuronal energy imbalance


Factors Impaired LEPRb signaling
Lepn Transport Defects
Genec Factors

HIF-α
Lepn Deleon of
Deficiency MC3R or MC4R
Fat
Insulin
Dysregulaon or
Resistance Lepn Increased Circulatory P53 Phosphorylaon of Rb
Lipodystrophy
Resistance Lepn

Obesity Nitric Oxide Apoptosis


ER Stress EPCs E2F
Chronic Inflammaon +
Feedback Angiogenesis &
Loop
Vasculogenesis
Hypertension

Melanoma
Diurec Use Cells

BCC
Proliferaon

Tumor Growth
Fig. 4 Potential interactions between leptin, basal cell cancer, and melanoma cells
Rev Endocr Metab Disord (2016) 17:389–403 395

[194]. The activation of this pathway also mediates melanoma obesity are endocrine distributing chemicals like
cell resistance to chemotherapeutic drugs. polychlorinated biphenyls [204], which are also associated
Inappropriately increased insulin and IGF are associated with the development of various cancers [205]. Many addi-
with inflammation, decreased physical activity, and an im- tives and preservatives like pesticides, herbicides, dyes, organ-
paired immune system [76]. IGF-1, in particular, associates ic amines, and polychlorinated phenyls bind melanin [62,
with VEGF to induce neovascularization and metastases 114–118]. This increases the pro-oxidant response of melanin,
[76]. Furthermore, IGF-2 is associated with tumorigenesis resulting in a build-up of ROS, which is a prominent feature
via the insulin and IGF receptors [76]. during melanoma pathogenesis [62]. Thus, quality of food
Serum levels of IGF-1 are significantly increased in pa- ingested can contribute to obesity as well as constitutive oxi-
tients with melanoma [195] in which lowered insulin-like- dative stress and melanoma [206].
factor-binding-proteins 3 and 5 (IGFBP-3 and IGFBP-5)
levels are associated with melanoma cell proliferation, metas-
tases, and reduced survival rates [36, 196]. 4.3.6 Medications

4.3.3 Glucocorticoids Some medications are associated with both obesity and pho-
tosensitization. Obesity also reduces the rate of drug metabo-
Glucocorticoids are the most common treatment for inflam- lism, increasing the intensity and duration of photosensitiza-
matory disorders including those of the skin; however, they tion [207], which could contribute to melanoma development.
also have significant negative effects on metabolism [197]. In Diuretics are the first line of treatment for individuals with
recurrent malignant melanoma patients, the glucocorticoid re- hypertension [208], and obesity and increased leptin levels
ceptor is overexpressed on melanoma cells [198]. facilitate hypertension. Overweight and obese individuals
Glucocorticoids have been shown to significantly reduce tu- using diuretics have an increased risk of BCC [209–212] be-
mor size and improve outcomes because they induce apopto- cause diuretics increase the risk of phototoxicity and
sis in melanoma cells [199]. photocarcinogenesis [213–215].
In obese individuals, glucocorticoids and psychological Obese individuals, because of chronic systemic inflamma-
stress also have an impact on appetite regulation and metabo- tion, may also take medications like nonsteroidal anti-
lism [200]. Obese individuals tend to have overactive 5-alpha- inflammatory drugs (NSAIDs), which are also associated with
reductase, which inactivates cortisol and, therefore, affects heightened photosensitivity [216, 217]. These cutaneous reac-
glucocorticoid metabolism [201]. tions could be due to the drugs’ effects on the immune system
via inflammatory pathways.
4.3.4 FTO gene SNPs BComfort eating^ is associated with psychological stress
and depressive symptoms [218], and psychiatric or mood-
More recently, it has been discovered that peroxisome stabilizing medications are known to increase patients’ sus-
proliferator-activated receptor gamma (PPARG) co-activators, ceptibility to UV damage [219–221]. Sugar and sweet taste
which are key regulators of energy metabolism, also regulate may have analgesic effects. Amazingly, an American child
the production of melanin in response to alpha-MSH in mela- after age 2 years on any given day is more likely to consume
nocytes. PPARG co-activators are also associated with tan- a sugar-sweetened product than a fruit or vegetable [222].
ning, providing a novel link between pigment formation and
energy metabolism [202].
The fat mass and obesity-associated protein (FTO) gene is
important in pigmentation and melanoma development [27].
While obesity-related SNPs are independent of both pigmen-
tary and melanoma-related FTO gene variants, it is likely that
the FTO gene influences multiple phenotypes. Common var-
iability in the FTO gene is associated with increased obesity
[203], and the FTO gene may also be important in pigmenta-
tion and melanoma development [27].

4.3.5 Diet

There are a variety of chemicals and environmental factors in Fig. 5 Basal cell cancer. This 16 × 10 mm ulcerated, crusted plaque with
food that contribute to the development of and rise in obesity a pearly border is noted on the left bridge of the nose in an elderly patient
worldwide. One particular group of chemicals linked to with sun-damaged skin
396 Rev Endocr Metab Disord (2016) 17:389–403

Table 4 Other possible risk factors for the development of non-


melanoma skin cancer

Diuretics or other photosensitizing medications [209–212]


Alcohol consumption [233]
High-fat diet [228]
Smoking [235]

Other risk factors of NMSC are noted in Table 3. Less well-


accepted risk factors are highlighted in Table 4. Obesity is best
characterized in this second group.
Fig. 6 Squamous cell cancer. A 6 × 6 cm eroded, crusted ulceration with In addition to the photosensitizing and mutagenic ef-
focal verrucous changes is noted on the right cheek and a 3 × 4 cm moist fects of acetaldehyde, alcohol consumption may promote
erythematous ulceration is noted on the right ear in this elderly patient skin cancer through impairment of the immune system
with a long history of sun exposure
compounded by poor nutritional status [236, 237]. In
fact, people who drink over 10 g/day of alcohol are 1.8
5 Non-melanoma skin cancers (NMSC)
times more likely to develop skin cancer than those who
do not drink alcohol [236, 237]. Alcohol consumption
Basal cell cancer (Fig. 5) originates from stem cells in the
may also be a marker for health risk behaviors like ex-
hair follicle infundibulum or interfollicular epidermis and
cess sun exposure [105–109]. Finally, cigarette smoke,
accounts for 90 % of all skin cancers [223]. It is primarily
which contains chemicals like arsenic, may be associated
driven by the sonic hedgehog pathway [224]. In the USA,
with cancers including SCC and BCC [235, 238–241].
approximately 1 million new cases of BCC are diagnosed
Long-term exposure to high levels of inorganic arsenic,
each year [225]. This rapid increase is likely due to life-
which occurs naturally but is also found due to pesticide and
style factors and levels of UVA radiation. Skin aging,
fertilizer use, is associated with higher rates of skin, bladder,
which primarily affects the basal cell layer of skin, is
and lung cancers, as well as heart disease. The FDA is currently
caused by both genetic and environmental factors and
examining these and other long-term effects and has set the
contributes to changes in DNA repair and stability, cell
maximum level of inorganic arsenic in infant rice cereal at
cycle and apoptosis, and cellular metabolism; aging may
1 0 0 - p a r t s - p e r - b i l l i o n ( h t t p : / / w w w. f d a .
also alter hormones that regulate tumor suppressor path-
gov/Food/FoodborneIllnessContaminants/Metals/ucm280202.
ways to influence carcinogenesis [226]. Squamous cell
htm).
carcinoma (Fig. 6) arises from the cells of the spinous
Factors that decrease the risk of developing NMSC are
layer of the epidermis. The incidence of squamous cell
detailed in Table 5.
carcinoma (SCC) has also drastically increased over the
past several decades [227]. A high-fat diet may contribute
to the development of SCC due to higher phospholipid
and cholesterol content in cancerous and pre-cancerous
6 Conclusion
skin lesions [228]. On the other hand, stromal adipocytes
may promote the differentiation of squamous cell carcino-
The dramatic increases in incidence of both obesity and skin
ma cells and inhibit their growth [229].
cancer emphasize the need to better understand the pathophys-
iology of both conditions and their connections. Melanoma is
considered the fastest growing cancer and non-melanoma skin
Table 3 Well-known risk factors for the development of non- cancer is also on the rise. The majority of the literature sug-
melanoma skin cancer
gests an association between obesity and both melanoma and
Ultraviolet radiation [12, 225, 230] NMSC. With several points of convergence, further research
Closer proximity to equator [231]
Age [232, 233] Table 5 Well-known factors that decrease risk for the development of
non-melanoma skin cancer
Fair eye color (blue, green, and hazel) [209]
Light hair color (red > blond > light brown) [209] Excess body weight [12, 13]
Fair complexion [209] Less baseline sun exposure [12, 13, 19, 242]
Immunosuppression [234] Less physical activity outdoors [12, 13, 19, 242]
Genetic influences [234] Less revealing clothing [233]
Rev Endocr Metab Disord (2016) 17:389–403 397

is necessary to explore and dissect the intricate relationships 15. Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-
mass index and incidence of cancer: a systematic review and meta-
between obesity and skin cancer.
analysis of prospective observational studies. Lancet.
2008;371(9612):569–78. doi:10.1016/S0140-6736(08)60269-X.
Compliance with ethical standards 16. Dobbins M, Decorby K, Choi BC. The association between obe-
sity and cancer risk: a meta-analysis of observational studies from
Conflict of interest The authors Karen Karimi, Robert Brodell, Taylor 1985 to 2011. ISRN Prev Med. 2013;2013:680536. doi:10.5402
Lindgren, and Christian A. Koch declare that there is no relevant conflict /2013/680536.
related to this work. 17. Veierod MB, Thelle DS, Laake P. Diet and risk of cutaneous ma-
lignant melanoma: a prospective study of 50,757 Norwegian men
and women. Int J Cancer. 1997;71(4):600–4.
18. Olsen CM, Green AC, Zens MS, Stukel TA, Bataille V, Berwick
M, et al. Anthropometric factors and risk of melanoma in women:
References a pooled analysis. Int J Cancer. 2008;122(5):1100–8. doi:10.1002
/ijc.23214.
1. Wolin KY, Carson K, Colditz GA. Obesity and cancer. Oncologist. 19. Praestegaard C, Kjaer SK, Christensen J, Tjonneland A, Halkjaer
2010;15(6):556–65. doi:10.1634/theoncologist.2009-0285. J, Jensen A. Obesity and risks for malignant melanoma and non-
2. Gallagher E, LeRoith D. Obesity and diabetes: the increased risk melanoma skin cancer: results from a large Danish prospective
of cancer and cancer-related mortality. Physiol Rev. 2015;95(3): cohort study. J Invest Dermatol. 2015;135(3):901–4. doi:10.1038
727–48. /jid.2014.438.
3. Albanes D, Winick M. Are cell number and cell proliferation risk 20. Fruhbeck G. Intracellular signalling pathways activated by leptin.
factors for cancer? J Natl Cancer Inst. 1988;80(10):772–4. Biochem J. 2006;393(Pt 1):7–20. doi:10.1042/BJ20051578.
4. Scrimshaw NS, SanGiovanni JP. Synergism of nutrition, infection, 21. Courneya KS, Katzmarzyk PT, Bacon E. Physical activity and
and immunity: an overview. Am J Clin Nutr. 1997;66(2):464S– obesity in Canadian cancer survivors: population-based estimates
from the 2005 Canadian Community Health Survey. Cancer.
77S.
2008;112(11):2475–82. doi:10.1002/cncr.23455.
5. Koch CA, Diamanti-Kandarakis E. Introduction to endocrine
22. Sahl WJ, Glore S, Garrison P, Oakleaf K, Johnson SD. Basal cell
disrupting chemicals—is it time to act? Rev Endocr Metab
carcinoma and lifestyle characteristics. Int J Dermatol.
Disord. 2015;16(4):269–70.
1995;34(6):398–402.
6. Kirkpatrick CS, White E, Lee JA. Case–control study of malig-
23. Flegal KM, Carroll MD, Ogden CL, Johnson CL. Prevalence and
nant melanoma in Washington State. II. Diet, alcohol, and obesity.
trends in obesity among US adults, 1999–2000. JAMA.
Am J Epidemiol. 1994;139(9):869–80.
2002;288(14):1723–7.
7. Samanic C, Chow WH, Gridley G, Jarvholm B, Fraumeni Jr JF. 24. Brandon EL, Gu JW, Cantwell L, He Z, Wallace G, Hall JE.
Relation of body mass index to cancer risk in 362,552 Swedish Obesity promotes melanoma tumor growth: role of leptin.
men. Cancer Causes Control. 2006;17(7):901–9. doi:10.1007 Cancer Biol Ther. 2009;8(19):1871–9.
/s10552-006-0023-9. 25. Monteiro CA, Moura EC, Conde WL, Popkin BM.
8. Samanic C, Gridley G, Chow WH, Lubin J, Hoover RN, Socioeconomic status and obesity in adult populations of devel-
Fraumeni Jr JF. Obesity and cancer risk among White and Black oping countries: a review. Bull World Health Organ.
United States veterans. Cancer Causes Control. 2004;15(1):35– 2004;82(12):940–6.
43. doi:10.1023/B:CACO.0000016573.79453.ba. 26. Popkin BM, Gordon-Larsen P. The nutrition transition: worldwide
9. Dubin N, Moseson M, Pasternack BS. Epidemiology of malignant obesity dynamics and their determinants. Int J Obes Relat Metab
melanoma: pigmentary traits, ultraviolet radiation, and the identi- Disord. 2004;28 Suppl 3:S2–9. doi:10.1038/sj.ijo.0802804.
fication of high-risk populations. Recent Results Cancer Res. 27. Li X, Liang L, Zhang M, Song F, Nan H, Wang LE, et al. Obesity-
1986;102:56–75. related genetic variants, human pigmentation, and risk of melano-
10. Gallus S, Naldi L, Martin L, Martinelli M, La Vecchia C. ma. Hum Genet. 2013;132(7):793–801. doi:10.1007/s00439-013-
Oncology Study Group of the Italian Group for Epidemiologic 1293-4.
Research in D. Anthropometric measures and risk of cutaneous 28. Frisancho AR, Wainwright R, Way A. Heritability and compo-
malignant melanoma: a case–control study from Italy. Melanoma nents of phenotypic expression in skin reflectance of Mestizos
Res. 2006;16(1):83–7. doi:10.1097/01. from the Peruvian lowlands. Am J Phys Anthropol. 1981;55(2):
cmr.0000194429.77643.76. 203–8. doi:10.1002/ajpa.1330550207.
11. Dennis LK, Lowe JB, Lynch CF, Alavanja MC. Cutaneous mela- 29. Harrison GA, Owen JJ. Studies on the inheritance of human skin
noma and obesity in the Agricultural Health Study. Ann colour. Ann Hum Genet. 1964;28:27–37.
Epidemiol. 2008;18(3):214–21. doi:10.1016/j. 30. Turner JB, Kumar A, Koch CA. The effects of indoor and outdoor
annepidem.2007.09.003. temperature on metabolic rate and adipose tissue—the Mississippi
12. Tang JY, Henderson MT, Hernandez-Boussard T, Kubo J, Desai perspective on the obesity epidemic. Rev Endocr Metab Disord.
M, Sims ST, et al. Lower skin cancer risk in women with higher 2016;17(1):61–71. doi:10.1007/s11154-016-9358-z.
body mass index: the women’s health initiative observational 31. Tai N, Wong FS, Wen L. The role of gut microbiota in the devel-
study. Cancer Epidemiol Biomarkers Prev. 2013;22(12):2412–5. opment of type 1, type 2 diabetes mellitus and obesity. Rev Endocr
doi:10.1158/1055-9965.EPI-13-0647. Metab Disord. 2015;16(1):55–65. doi:10.1007/s11154-015-9309-
13. Pothiawala S, Qureshi AA, Li Y, Han J. Obesity and the incidence 0.
of skin cancer in US Caucasians. Cancer Causes Control : CCC. 32. Gerstenblith MR, Shi J, Landi MT. Genome-wide association
2012;23(5):717–26. doi:10.1007/s10552-012-9941-x. studies of pigmentation and skin cancer: a review and meta-anal-
14. Sergentanis TN, Antoniadis AG, Gogas HJ, Antonopoulos CN, ysis. Pigment Cell Melanoma Res. 2010;23(5):587–606.
Adami HO, Ekbom A, et al. Obesity and risk of malignant mela- doi:10.1111/j.1755-148X.2010.00730.x.
noma: a meta-analysis of cohort and case–control studies. Eur J 33. Nan H, Kraft P, Hunter DJ, Han J. Genetic variants in pigmenta-
Cancer. 2013;49(3):642–57. doi:10.1016/j.ejca.2012.08.028. tion genes, pigmentary phenotypes, and risk of skin cancer in
398 Rev Endocr Metab Disord (2016) 17:389–403

Caucasians. Int J Cancer. 2009;125(4):909–17. doi:10.1002 carcinogenic risk associated with the intake of multiple relevant
/ijc.24327. carcinogens found in meat and charcuterie products. Sci Total
34. McAllister EJ, Dhurandhar NV, Keith SW, Aronne LJ, Barger J, Environ. 2015;514:33–41. doi:10.1016/j.scitotenv.2015.01.108.
Baskin M, et al. Ten putative contributors to the obesity epidemic. 52. Schut HA, Snyderwine EG. DNA adducts of heterocyclic amine
Crit Rev Food Sci Nutr. 2009;49(10):868–913. doi:10.1080 food mutagens: implications for mutagenesis and carcinogenesis.
/10408390903372599. Carcinogenesis. 1999;20(3):353–68.
35. Charriere G, Cousin B, Arnaud E, Andre M, Bacou F, Penicaud L, 53. Pei YF, Tian Q, Zhang L, Deng HW. Exploring the major sources
et al. Preadipocyte conversion to macrophage. Evidence of plas- and extent of heterogeneity in a genome-wide association meta-
ticity. J Biol Chem. 2003;278(11):9850–5. doi:10.1074/jbc. analysis. Ann Hum Genet. 2015. doi:10.1111/ahg.12143.
M210811200. 54. Holmes L, LaHurd A, Wasson E, McClarin L, Dabney K. Racial
36. Wang J, Ding N, Li Y, Cheng H, Wang D, Yang Q, et al. Insulin- and ethnic heterogeneity in the association between total choles-
like growth factor binding protein 5 (IGFBP5) functions as a tu- terol and pediatric obesity. Int J Environ Res Public Health.
mor suppressor in human melanoma cells. Oncotarget. 2015;13(1). doi:10.3390/ijerph13010019.
2015;6(24):20636–49. 55. Perl A. mTOR activation is a biomarker and a central pathway to
37. Wang H, Ye J. Regulation of energy balance by inflammation: autoimmune disorders, cancer, obesity, and aging. Ann N Y Acad
common theme in physiology and pathology. Rev Endocr Metab Sci. 2015;1346(1):33–44. doi:10.1111/nyas.12756.
Disord. 2015;16(1):47–54. 56. Cowen S, McLaughlin SL, Hobbs G, Coad J, Martin KH, Olfert
38. Kloting N, Bluher M. Adipocyte dysfunction, inflammation and IM, et al. High-fat, high-calorie diet enhances mammary carcino-
metabolic syndrome. Rev Endocr Metab Disord. 2014;15(4):277– genesis and local inflammation in MMTV-PyMT mouse model of
87. breast cancer. Cancers (Basel). 2015;7(3):1125–42. doi:10.3390
39. Ngo HT, Hetland RB, Nygaard UC, Steffensen IL. Genetic and /cancers7030828.
diet-induced obesity increased intestinal tumorigenesis in the dou- 57. Crujeiras AB, Cabia B, Carreira MC, Amil M, Cueva J, Andrade
ble mutant mouse model multiple intestinal neoplasia X obese via S, et al. Secreted factors derived from obese visceral adipose tissue
disturbed glucose regulation and inflammation. J Obes. regulate the expression of breast malignant transformation genes.
2015;2015:343479. doi:10.1155/2015/343479. Int J Obes (Lond). 2015. doi:10.1038/ijo.2015.208.
40. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic
58. Gorlach A, Dimova EY, Petry A, Martinez-Ruiz A, Hernansanz-
switch in adipose tissue macrophage polarization. J Clin Invest.
Agustin P, Rolo AP, et al. Reactive oxygen species, nutrition,
2007;117(1):175–84. doi:10.1172/JCI29881.
hypoxia and diseases: problems solved? Redox Biol. 2015;6:
41. Nguyen MT, Favelyukis S, Nguyen AK, Reichart D, Scott PA,
372–85. doi:10.1016/j.redox.2015.08.016.
Jenn A, et al. A subpopulation of macrophages infiltrates hyper-
59. Hasan S, Suhail N, Bilal N, Ashraf GM, Zaidi SK, AlNohair S,
trophic adipose tissue and is activated by free fatty acids via Toll-
et al. Chronic unpredictable stress deteriorates the chemopreven-
like receptors 2 and 4 and JNK-dependent pathways. J Biol Chem.
tive efficacy of pomegranate through oxidative stress pathway.
2007;282(48):35279–92. doi:10.1074/jbc.M706762200.
Tumour Biol. 2015. doi:10.1007/s13277-015-4469-9.
42. Strissel KJ, Stancheva Z, Miyoshi H, Perfield 2nd JW, DeFuria J,
60. Manna P, Jain SK. Obesity, oxidative stress, adipose tissue dys-
Jick Z, et al. Adipocyte death, adipose tissue remodeling, and
function, and the associated health risks: causes and therapeutic
obesity complications. Diabetes. 2007;56(12):2910–8.
strategies. Metab Syndr Relat Disord. 2015;13(10):423–44.
doi:10.2337/db07-0767.
doi:10.1089/met.2015.0095.
43. Zaldivar F, McMurray RG, Nemet D, Galassetti P, Mills PJ,
Cooper DM. Body fat and circulating leukocytes in children. Int 61. Hunsche C, Hernandez O, De la Fuente M. Impaired immune
J Obes (Lond). 2006;30(6):906–11. doi:10.1038/sj.ijo.0803227. response in old mice suffering from obesity and premature
44. Debnath M, Agrawal S, Agrawal A, Dubey GP. Metaflammatory immunosenescence in adulthood. J Gerontol A Biol Sci Med
responses during obesity: pathomechanism and treatment. Obes Sci. 2015. doi:10.1093/gerona/glv082.
Res Clin Pract. 2015. doi:10.1016/j.orcp.2015.10.012. 62. Meyskens Jr FL, Farmer PJ, Anton-Culver H. Etiologic pathogen-
45. Lumeng CN, Saltiel AR. Inflammatory links between obesity and esis of melanoma: a unifying hypothesis for the missing attribut-
metabolic disease. J Clin Invest. 2011;121(6):2111–7. doi:10.1172 able risk. Clin Cancer Res : Off J Am Assoc Cancer Res.
/JCI57132. 2004;10(8):2581–3.
46. Shore SA. Environmental perturbations: obesity. Compr Physiol. 63. Caslin HL, Franco RL, Crabb EB, Huang CJ, Bowen MK,
2011;1(1):263–82. doi:10.1002/cphy.c100017. Acevedo EO. The effect of obesity on inflammatory cytokine
47. Atabek ME, Vatansev H, Erkul I. Oxidative stress in childhood and leptin production following acute mental stress.
obesity. J Pediatr Endocrinol Metab. 2004;17(8):1063–8. Psychophysiology. 2015. doi:10.1111/psyp.12568.
48. Bakker SJ RGIJ, Teerlink T, Westerhoff HV, Gans RO, Heine RJ. 64. Gogas H, Trakatelli M, Dessypris N, Terzidis A, Katsambas A,
Cytosolic triglycerides and oxidative stress in central obesity: the Chrousos GP, et al. Melanoma risk in association with serum
missing link between excessive atherosclerosis, endothelial dys- leptin levels and lifestyle parameters: a case–control study. Ann
function, and beta-cell failure? Atherosclerosis. 2000;148(1):17– Oncol. 2008;19(2):384–9. doi:10.1093/annonc/mdm464.
21. 65. Dayakar A, Chandrasekaran S, Veronica J, Maurya RS. Leptin
49. Keaney Jr JF, Larson MG, Vasan RS, Wilson PW, Lipinska I, induces the phagocytosis and protective immune response in
Corey D, et al. Obesity and systemic oxidative stress: clinical Leishmania donovani infected THP-1 cell line and human
correlates of oxidative stress in the Framingham Study. PBMCs. Exp Parasitol. 2015. doi:10.1016/j.exppara.2015.12.002.
Arterioscler Thromb Vasc Biol. 2003;23(3):434–9. doi:10.1161 66. Huang CJ, McAllister MJ, Slusher AL, Webb HE, Mock JT,
/01.ATV.0000058402.34138.11. Acevedo EO. Obesity-related oxidative stress: the impact of phys-
50. Lee YS, Kim AY, Choi JW, Kim M, Yasue S, Son HJ, et al. ical activity and diet manipulation. Sports Med Open. 2015;1(1):
Dysregulation of adipose glutathione peroxidase 3 in obesity con- 32. doi:10.1186/s40798-015-0031-y.
tributes to local and systemic oxidative stress. Mol Endocrinol. 67. Mizutani H, Fukushima S, Masuguchi S, Yamashita J, Miyashita
2008;22(9):2176–89. doi:10.1210/me.2008-0023. A, Nakahara S, et al. Serum levels of leptin receptor in patients
51. Hernandez AR, Boada LD, Almeida-Gonzalez M, Mendoza Z, with malignant melanoma as a new tumor marker. Exp Dermatol.
Ruiz-Suarez N, Valeron PF, et al. An estimation of the 2013;22(11):748–9. doi:10.1111/exd.12238.
Rev Endocr Metab Disord (2016) 17:389–403 399

68. Chirumbolo S. The role of vitamin D towards immune tolerance in 88. Pollitt RA, Clarke CA, Shema SJ, Swetter SM. California
white adipose tissue (WAT). Endocr Metab Immune Disord Drug Medicaid enrollment and melanoma stage at diagnosis: a
Targets. 2015;15(4):277–87. population-based study. Am J Prev Med. 2008;35(1):7–13.
69. Yao Y, Zhu L, He L, Duan Y, Liang W, Nie Z, et al. A meta- doi:10.1016/j.amepre.2008.03.026.
analysis of the relationship between vitamin D deficiency and 89. Roetzheim RG, Pal N, Tennant C, Voti L, Ayanian JZ, Schwabe A,
obesity. Int J Clin Exp Med. 2015;8(9):14977–84. et al. Effects of health insurance and race on early detection of
70. Lagunova Z, Porojnicu AC, Grant WB, Bruland O, Moan JE. cancer. J Natl Cancer Inst. 1999;91(16):1409–15.
Obesity and increased risk of cancer: does decrease of serum 25- 90. Diffey B. Climate change, ozone depletion and the impact on
hydroxyvitamin D level with increasing body mass index explain ultraviolet exposure of human skin. Phys Med Biol.
some of the association? Mol Nutr Food Res. 2010;54(8):1127– 2004;49(1):R1–11.
33. doi:10.1002/mnfr.200900512. 91. Welch HG, Woloshin S, Schwartz LM. Skin biopsy rates and
71. Kremer R, Campbell PP, Reinhardt T, Gilsanz V. Vitamin D status incidence of melanoma: population based ecological study.
and its relationship to body fat, final height, and peak bone mass in BMJ. 2005;331(7515):481. doi:10.1136/bmj.38516.649537.E0.
young women. J Clin Endocrinol Metab. 2009;94(1):67–73. 92. Hall HI, Jamison P, Fulton JP, Clutter G, Roffers S, Parrish P.
doi:10.1210/jc.2008-1575. Reporting cutaneous melanoma to cancer registries in the United
72. Bikle DD. Vitamin D, and the skin: physiology and pathophysi- States. J Am Acad Dermatol. 2003;49(4):624–30.
ology. Rev Endocr Metab Disord. 2012;13(1):3–19. 93. Swerlick RA, Chen S. The melanoma epidemic. Is increased sur-
73. Holick MF. Vitamin D: importance in the prevention of cancers, veillance the solution or the problem? Arch Dermatol.
type 1 diabetes, heart disease, and osteoporosis. Am J Clin Nutr. 1996;132(8):881–4.
2004;79(3):362–71. 94. Swerlick RA, Chen S. The melanoma epidemic: more apparent
74. Lackey DE, Olefsky JM. Regulation of metabolism by the innate than real? Mayo Clin Proc. 1997;72(6):559–64. doi:10.1016
immune system. Nat Rev Endocrinol. 2016;12(1):15–28. /S0025-6196(11)63306-5.
doi:10.1038/nrendo.2015.189. 95. Olsen CM, Carroll HJ, Whiteman DC. Estimating the attributable
75. Zeyda M, Stulnig TM. Adipose tissue macrophages. Immunol fraction for cancer: a meta-analysis of nevi and melanoma. Cancer
Lett. 2007;112(2):61–7. doi:10.1016/j.imlet.2007.07.003. Prev Res (Phila). 2010;3(2):233–45. doi:10.1158/1940-6207.
76. De Pergola G, Silvestris F. Obesity as a major risk factor for CAPR-09-0108.
cancer. J Obes. 2013;2013:291546. doi:10.1155/2013/291546. 96. Gandini S, Sera F, Cattaruzza MS, Pasquini P, Zanetti R, Masini C,
77. Saraiya A, Al-Shoha A, Brodell RT. Hyperinsulinemia associated et al. Meta-analysis of risk factors for cutaneous melanoma: III.
with acanthosis nigricans, finger pebbles, acrochordons, and the Family history, actinic damage and phenotypic factors. Eur J
sign of Leser-Trelat. Endocr Pract. 2013;19(3):522–5. Cancer. 2005;41(14):2040–59. doi:10.1016/j.ejca.2005.03.034.
doi:10.4158/EP12192.RA. 97. Wheless L, Black J, Alberg AJ. Nonmelanoma skin cancer and the
78. East HE, Subauste JS, Gandhi A, Koch CA. About secondary risk of second primary cancers: a systematic review. Cancer
causes of diabetes mellitus. J Miss State Med Assoc. Epidemiol Biomarkers Prev. 2010;19(7):1686–95. doi:10.1158
2012;53(11):380–3. /1055-9965.EPI-10-0243.
79. Cheng S, Gomez K, Serri O, Chik C, Ezzat S. The role of diabetes 98. Chen ST, Geller AC, Tsao H. Update on the epidemiology of
in acromegaly associated neoplasia. PLoS One. 2015;10(5), melanoma. Curr Dermatol Rep. 2013;2(1):24–34. doi:10.1007
e0127276. doi:10.1371/journal.pone.0127276. /s13671-012-0035-5.
80. Su Y, Carey LC, Rose JC, Pulgar VM. Leptin alters adrenal re- 99. Pappo AS, Armstrong GT, Liu W, Srivastava DK, McDonald A,
sponsiveness by decreasing expression of ACTH-R, StAR, and Leisenring WM, et al. Melanoma as a subsequent neoplasm in
P450c21 in hypoxemic fetal sheep. Reprod Sci. 2012;19(10): adult survivors of childhood cancer: a report from the childhood
1075–84. doi:10.1177/1933719112442246. cancer survivor study. Pediatr Blood Cancer. 2013;60(3):461–6.
81. Ellerhorst JA, Diwan AH, Dang SM, Uffort DG, Johnson MK, doi:10.1002/pbc.24266.
Cooke CP, et al. Promotion of melanoma growth by the metabolic 100. Gandini S, Sera F, Cattaruzza MS, Pasquini P, Picconi O, Boyle P,
hormone leptin. Oncol Rep. 2010;23(4):901–7. et al. Meta-analysis of risk factors for cutaneous melanoma: II.
82. de Vries E, Coebergh JW. Cutaneous malignant melanoma in Sun exposure. Eur J Cancer. 2005;41(1):45–60. doi:10.1016/j.
Europe. Eur J Cancer. 2004;40(16):2355–66. doi:10.1016/j. ejca.2004.10.016.
ejca.2004.06.003. 101. Rigel DS, Rigel EG, Rigel AC. Effects of altitude and latitude on
83. Lasithiotakis KG, Leiter U, Gorkievicz R, Eigentler T, Breuninger ambient UVB radiation. J Am Acad Dermatol. 1999;40(1):114–6.
H, Metzler G, et al. The incidence and mortality of cutaneous 102. Bulliard JL. Site-specific risk of cutaneous malignant melanoma
melanoma in Southern Germany: trends by anatomic site and and pattern of sun exposure in New Zealand. Int J Cancer.
pathologic characteristics, 1976 to 2003. Cancer. 2006;107(6): 2000;85(5):627–32.
1331–9. doi:10.1002/cncr.22126. 103. Parisi AV, Kimlin MG, Lester R, Turnbull D. Lower body ana-
84. Mansson-Brahme E, Johansson H, Larsson O, Rutqvist LE, tomical distribution of solar ultraviolet radiation on the human
Ringborg U. Trends in incidence of cutaneous malignant melano- form in standing and sitting postures. J Photochem Photobiol B.
ma in a Swedish population 1976–1994. Acta Oncol. 2002;41(2): 2003;69(1):1–6.
138–46. 104. Reed KB, Brewer JD, Lohse CM, Bringe KE, Pruitt CN, Gibson
85. Stang A, Pukkala E, Sankila R, Soderman B, Hakulinen T. Time LE. Increasing incidence of melanoma among young adults: an
trend analysis of the skin melanoma incidence of Finland from epidemiological study in Olmsted County. Minnesota Mayo Clin
1953 through 2003 including 16,414 cases. Int J Cancer. Proc. 2012;87(4):328–34. doi:10.1016/j.mayocp.2012.01.010.
2006;119(2):380–4. doi:10.1002/ijc.21836. 105. Stryker WS, Stampfer MJ, Stein EA, Kaplan L, Louis TA, Sober
86. Linos E, Swetter SM, Cockburn MG, Colditz GA, Clarke CA. A, et al. Diet, plasma levels of beta-carotene and alpha-tocopherol,
Increasing burden of melanoma in the United States. J Invest and risk of malignant melanoma. Am J Epidemiol. 1990;131(4):
Dermatol. 2009;129(7):1666–74. doi:10.1038/jid.2008.423. 597–611.
87. Ortiz CA, Goodwin JS, Freeman JL. The effect of socioeconomic 106. Bain C, Green A, Siskind V, Alexander J, Harvey P. Diet and
factors on incidence, stage at diagnosis and survival of cutaneous melanoma. An exploratory case–control study. Ann Epidemiol.
melanoma. Med Sci Monit. 2005;11(5):RA163–72. 1993;3(3):235–8.
400 Rev Endocr Metab Disord (2016) 17:389–403

107. Millen AE, Tucker MA, Hartge P, Halpern A, Elder DE, Guerry D, 128. D’Orazio JA, Nobuhisa T, Cui R, Arya M, Spry M, Wakamatsu K,
et al. Diet and melanoma in a case–control study. Cancer et al. Topical drug rescue strategy and skin protection based on the
Epidemiol Biomarkers Prev. 2004;13(6):1042–51. role of Mc1r in UV-induced tanning. Nature. 2006;443(7109):
108. Meyskens Jr FL, Farmer PJ, Anton-Culver H. Diet and melanoma 340–4. doi:10.1038/nature05098.
in a case–control study. Cancer Epidemiol Biomarkers Prev. 129. Cui R, Widlund HR, Feige E, Lin JY, Wilensky DL, Igras VE,
2005;14(1):293. et al. Central role of p53 in the suntan response and pathologic
109. Le Marchand L, Saltzman BS, Hankin JH, Wilkens LR, Franke hyperpigmentation. Cell. 2007;128(5):853–64. doi:10.1016/j.
AA, Morris SJ, et al. Sun exposure, diet, and melanoma in Hawaii cell.2006.12.045.
Caucasians. Am J Epidemiol. 2006;164(3):232–45. doi:10.1093 130. Sato HNY, Chrousos GP, Ichihashi M, Funasak Y. The expression
/aje/kwj115. of corticotropin-releasing hormone in melanoma. Pigment Cell
110. Nicolopoulou-Stamati P, Hens L, Sasco AJ. Cosmetics as endo- Res. 2002;15(2):98–103.
crine disruptors: are they a health risk? Rev Endocr Metab Disord. 131. Millington GW. The role of proopiomelanocortin (POMC)
2015;16(4):373–83. neurones in feeding behaviour. Nutr Metab (Lond). 2007;4:18.
111. Holland KT, Bojar RA. Cosmetics: what is their influence on the doi:10.1186/1743-7075-4-18.
skin microflora? Am J Clin Dermatol. 2002;3(7):445–9. 132. Sharma SD, Katiyar SK. Leptin deficiency-induced obesity exac-
112. Karpuzoglu E, Holladay SD, Gogal Jr RM. Parabens: potential erbates ultraviolet B radiation-induced cyclooxygenase-2 expres-
impact of low-affinity estrogen receptor binding chemicals on hu- sion and cell survival signals in ultraviolet B-irradiated mouse
man health. J Toxicol Environ Health B Crit Rev. 2013;16(5):321– skin. Toxicol Appl Pharmacol. 2010;244(3):328–35. doi:10.1016
35. doi:10.1080/10937404.2013.809252. /j.taap.2010.01.010.
113. Gandini S, Iodice S, Koomen E, Di Pietro A, Sera F, Caini S. 133. Akbani R, Akdemir KC, Aksoy BA, Albert M, Ally A, Amin SB,
Hormonal and reproductive factors in relation to melanoma in et al. Genomic classification of cutaneous melanoma. Cell.
women: current review and meta-analysis. Eur J Cancer. 2015;161(7):1681–96. doi:10.1016/j.cell.2015.05.044.
2011;47(17):2607–17. doi:10.1016/j.ejca.2011.04.023. 134. Garofalo C, Surmacz E. Leptin and cancer. J Cell Physiol.
114. Ward EM, Burnett CA, Ruder A, Davis-King K. Industries and 2006;207(1):12–22. doi:10.1002/jcp.20472.
cancer. Cancer Causes Control. 1997;8(3):356–70. 135. Tessitore L, Vizio B, Jenkins O, De Stefano I, Ritossa C, Argiles
115. Sinks T, Steele G, Smith AB, Watkins K, Shults RA. Mortality JM, et al. Leptin expression in colorectal and breast cancer pa-
among workers exposed to polychlorinated biphenyls. Am J tients. Int J Mol Med. 2000;5(4):421–6.
Epidemiol. 1992;136(4):389–98.
136. Petridou E, Belechri M, Dessypris N, Koukoulomatis P,
116. Loomis D, Browning SR, Schenck AP, Gregory E, Savitz DA.
Diakomanolis E, Spanos E, et al. Leptin and body mass index
Cancer mortality among electric utility workers exposed to
in relation to endometrial cancer risk. Ann Nutr Metab.
polychlorinated biphenyls. Occup Environ Med. 1997;54(10):
2002;46(3–4):147–51.
720–8.
137. Stattin P, Palmqvist R, Soderberg S, Biessy C, Ardnor B,
117. Robinson CF, Petersen M, Palu S. Mortality patterns among elec-
Hallmans G, et al. Plasma leptin and colorectal cancer risk: a
trical workers employed in the U.S. construction industry, 1982–
prospective study in Northern Sweden. Oncol Rep. 2003;10(6):
1987. Am J Ind Med. 1999;36(6):630–7.
2015–21.
118. Nelemans PJ, Scholte R, Groenendal H, Kiemeney LA, Rampen
FH, Ruiter DJ, et al. Melanoma and occupation: results of a case– 138. Arnardottir ES, Maislin G, Jackson N, Schwab RJ, Benediktsdottir
control study in The Netherlands. Br J Ind Med. 1993;50(7):642– B, Teff K, et al. The role of obesity, different fat compartments and
6. sleep apnea severity in circulating leptin levels: the Icelandic Sleep
119. Gantz I, Fong TM. The melanocortin system. Am J Physiol Apnea Cohort study. Int J Obes (Lond). 2013;37(6):835–42.
Endocrinol Metab. 2003;284(3):E468–74. doi:10.1152 doi:10.1038/ijo.2012.138.
/ajpendo.00434.2002. 139. Leroy P, Dessolin S, Villageois P, Moon BC, Friedman JM,
120. Danforth CH. Hereditary adiposity in mice. Obes Res. 1996;4(1): Ailhaud G, et al. Expression of ob gene in adipose cells.
96–100. Regulation by insulin. J Biol Chem. 1996;271(5):2365–8.
121. Dickies MM. A new viable yellow mutation in the house mouse. J 140. Zhang HH, Kumar S, Barnett AH, Eggo MC. Tumour necrosis
Hered. 1962;53:84–6. factor-alpha exerts dual effects on human adipose leptin synthesis
122. Heston WE, Vlahakis G. Influence of the Ay gene on mammary- and release. Mol Cell Endocrinol. 2000;159(1–2):79–88.
gland tumors, hepatomas, and normal growth in mice. J Natl 141. Dagogo-Jack S, Selke G, Melson AK, Newcomer JW. Robust
Cancer Inst. 1961;26:969–83. leptin secretory responses to dexamethasone in obese subjects. J
123. Heston WE, Vlahakis G. C3H-Avy—a high hepatoma and high Clin Endocrinol Metab. 1997;82(10):3230–3. doi:10.1210
mammary tumor strain of mice. J Natl Cancer Inst. 1968;40(6): /jcem.82.10.4154.
1161–6. 142. Machinal-Quelin F, Dieudonne MN, Pecquery R, Leneveu MC,
124. Yen TT, Gill AM, Frigeri LG, Barsh GS, Wolff GL. Obesity, Giudicelli Y. Direct in vitro effects of androgens and estrogens on
diabetes, and neoplasia in yellow A(vy)/- mice: ectopic expression ob gene expression and leptin secretion in human adipose tissue.
of the agouti gene. FASEB J. 1994;8(8):479–88. Endocrine. 2002;18(2):179–84. doi:10.1385/ENDO:18:2:179.
125. Krude H, Biebermann H, Luck W, Horn R, Brabant G, Gruters A. 143. Fain JN, Leffler CW, Bahouth SW. Eicosanoids as endogenous
Severe early-onset obesity, adrenal insufficiency and red hair pig- regulators of leptin release and lipolysis by mouse adipose tissue
mentation caused by POMC mutations in humans. Nat Genet. in primary culture. J Lipid Res. 2000;41(10):1689–94.
1998;19(2):155–7. doi:10.1038/509. 144. Oba J, Wei W, Gershenwald JE, Johnson MM, Wyatt CM,
126. Millington GW. Proopiomelanocortin (POMC): the cutaneous Ellerhorst JA, et al. Elevated serum leptin levels are associated
roles of its melanocortin products and receptors. Clin Exp with an increased risk of sentinel lymph node metastasis in cuta-
D e r m a t o l . 2 0 0 6 ; 3 1 ( 3 ) : 4 0 7 – 1 2 . d o i : 1 0 . 1111 / j . 1 3 6 5 - neous melanoma. Medicine (Baltimore). 2016;95(11), e3073.
2230.2006.02128.x. doi:10.1097/MD.0000000000003073.
127. Ramachandrappa S, Farooqi IS. Genetic approaches to under- 145. Jung JI, Cho HJ, Jung YJ, Kwon SH, Her S, Choi SS, et al. High-
standing human obesity. J Clin Invest. 2011;121(6):2080–6. fat diet-induced obesity increases lymphangiogenesis and lymph
doi:10.1172/JCI46044. node metastasis in the B16F10 melanoma allograft model: roles of
Rev Endocr Metab Disord (2016) 17:389–403 401

adipocytes and M2-macrophages. Int J Cancer. 2015;136(2):258– cardiovascular disease. J Am Coll Cardiol. 2008;52(15):1201–
70. doi:10.1002/ijc.28983. 10. doi:10.1016/j.jacc.2008.05.060.
146. McMurphy T, Xiao R, Magee D, Slater A, Zabeau L, Tavernier J, 164. Mantzoros CS, Liolios AD, Tritos NA, Kaklamani VG,
et al. The anti-tumor activity of a neutralizing nanobody targeting Doulgerakis DE, Griveas I, et al. Circulating insulin concentra-
leptin receptor in a mouse model of melanoma. PLoS ONE. tions, smoking, and alcohol intake are important independent pre-
2014;9(2), e89895. doi:10.1371/journal.pone.0089895. dictors of leptin in young healthy men. Obes Res. 1998;6(3):179–
147. Emilsson V, Liu YL, Cawthorne MA, Morton NM, Davenport M. 86.
Expression of the functional leptin receptor mRNA in pancreatic 165. Traish AM, Zitzmann M. The complex and multifactorial relation-
islets and direct inhibitory action of leptin on insulin secretion. ship between testosterone deficiency (TD), obesity and vascular
Diabetes. 1997;46(2):313–6. disease. Rev Endocr Metab Disord. 2015;16(3):249–68.
148. Kulkarni RN, Wang ZL, Wang RM, Hurley JD, Smith DM, Ghatei doi:10.1007/s11154-015-9323-2.
MA, et al. Leptin rapidly suppresses insulin release from 166. Ullah MI, Washington T, Kazi M, Tamanna S, Koch CA.
insulinoma cells, rat and human islets, and in vivo, in mice. J Testosterone deficiency as a risk factor for cardiovascular disease.
Clin Invest. 1997;100(11):2729–36. doi:10.1172/JCI119818. Horm Metab Res. 2011;43(03):153–64.
149. Morris DL, Rui L. Recent advances in understanding leptin sig- 167. Coppari R, Bjorbaek C. Leptin revisited: its mechanism of action
naling and leptin resistance. Am J Physiol Endocrinol Metab. and potential for treating diabetes. Nat Rev Drug Discov.
2009;297(6):E1247–59. doi:10.1152/ajpendo.00274.2009. 2012;11(9):692–708. doi:10.1038/nrd3757.
150. Wang MY, Lee Y, Unger RH. Novel form of lipolysis induced by 168. Stanley BG, Leibowitz SF. Neuropeptide Y injected in the
leptin. J Biol Chem. 1999;274(25):17541–4. paraventricular hypothalamus: a powerful stimulant of feeding
151. Campfield LA, Smith FJ, Guisez Y, Devos R, Burn P. behavior. Proc Natl Acad Sci U S A. 1985;82(11):3940–3.
Recombinant mouse OB protein: evidence for a peripheral signal 169. Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, et al.
linking adiposity and central neural networks. Science. Endoplasmic reticulum stress plays a central role in development
1995;269(5223):546–9. of leptin resistance. Cell Metab. 2009;9(1):35–51. doi:10.1016/j.
152. Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, cmet.2008.12.004.
Rabinowitz D, et al. Weight-reducing effects of the plasma protein 170. Zhang X, Zhang G, Zhang H, Karin M, Bai H, Cai D.
encoded by the obese gene. Science. 1995;269(5223):543–6. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutri-
153. Pelleymounter MA, Cullen MJ, Baker MB, Hecht R, Winters D, tion to energy imbalance and obesity. Cell. 2008;135(1):61–73.
Boone T, et al. Effects of the obese gene product on body weight doi:10.1016/j.cell.2008.07.043.
regulation in ob/ob mice. Science. 1995;269(5223):540–3. 171. Ogimoto K, Harris Jr MK, Wisse BE. MyD88 is a key mediator of
154. Stephens TW, Basinski M, Bristow PK, Bue-Valleskey JM, anorexia, but not weight loss, induced by lipopolysaccharide and
Burgett SG, Craft L, et al. The role of neuropeptide Y in the interleukin-1 beta. Endocrinology. 2006;147(9):4445–53.
antiobesity action of the obese gene product. Nature. doi:10.1210/en.2006-0465.
1995;377(6549):530–2. doi:10.1038/377530a0. 172. Ozcan U, Ozcan L, Yilmaz E, Duvel K, Sahin M, Manning BD,
155. Caro JF, Kolaczynski JW, Nyce MR, Ohannesian JP, Opentanova et al. Loss of the tuberous sclerosis complex tumor suppressors
I, Goldman WH, et al. Decreased cerebrospinal-fluid/serum leptin triggers the unfolded protein response to regulate insulin signaling
ratio in obesity: a possible mechanism for leptin resistance. and apoptosis. Mol Cell. 2008;29(5):541–51. doi:10.1016/j.
Lancet. 1996;348(9021):159–61. molcel.2007.12.023.
156. El-Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS. 173. Hosoi T, Sasaki M, Miyahara T, Hashimoto C, Matsuo S, Yoshii
Two defects contribute to hypothalamic leptin resistance in mice M, et al. Endoplasmic reticulum stress induces leptin resistance.
with diet-induced obesity. J Clin Invest. 2000;105(12):1827–32. M o l P h a r m a c o l . 2 0 0 8 ; 7 4 ( 6 ) : 1 6 1 0 – 9 . d o i : 1 0 . 11 2 4
doi:10.1172/JCI9842. /mol.108.050070.
157. Schwartz MW, Peskind E, Raskind M, Boyko EJ, Porte Jr D. 174. Lam QL, Lu L. Role of leptin in immunity. Cell Mol Immunol.
Cerebrospinal fluid leptin levels: relationship to plasma levels 2007;4(1):1–13.
and to adiposity in humans. Nat Med. 1996;2(5):589–93. 175. Chen K, Li F, Li J, Cai H, Strom S, Bisello A, et al. Induction of
158. Belouzard S, Delcroix D, Rouille Y. Low levels of expression leptin resistance through direct interaction of C-reactive protein
of leptin receptor at the cell surface result from constitutive with leptin. Nat Med. 2006;12(4):425–32. doi:10.1038/nm1372.
endocytosis and intracellular retention in the biosynthetic 176. Amjadi F, Javanmard SH, Zarkesh-Esfahani H, Khazaei M,
p a t h w a y. J B i o l C h e m . 2 0 0 4 ; 2 7 9 ( 2 7 ) : 2 8 4 9 9 – 5 0 8 . Narimani M. Leptin promotes melanoma tumor growth in mice
doi:10.1074/jbc.M400508200. related to increasing circulating endothelial progenitor cells num-
159. Diano S, Kalra SP, Horvath TL. Leptin receptor immunoreactivity bers and plasma NO production. J Exp Clin Cancer Res. 2011;30:
is associated with the Golgi apparatus of hypothalamic neurons 21. doi:10.1186/1756-9966-30-21.
and glial cells. J Neuroendocrinol. 1998;10(9):647–50. 177. Dome B, Hendrix MJ, Paku S, Tovari J, Timar J. Alternative
160. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, vascularization mechanisms in cancer: pathology and therapeutic
Wareham NJ, et al. Congenital leptin deficiency is associated with implications. Am J Pathol. 2007;170(1):1–15. doi:10.2353
severe early-onset obesity in humans. Nature. 1997;387(6636): /ajpath.2007.060302.
903–8. doi:10.1038/43185. 178. Gonzalez RR, Cherfils S, Escobar M, Yoo JH, Carino C, Styer
161. Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD. A leptin AK, et al. Leptin signaling promotes the growth of mammary
missense mutation associated with hypogonadism and morbid tumors and increases the expression of vascular endothelial
obesity. Nat Genet. 1998;18(3):213–5. doi:10.1038/ng0398-213. growth factor (VEGF) and its receptor type two (VEGF-R2). J
162. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman Biol Chem. 2006;281(36):26320–8. doi:10.1074/jbc.
JM. Positional cloning of the mouse obese gene and its human M601991200.
homologue. Nature. 1994;372(6505):425–32. doi:10.1038 179. Ribatti D, Nico B, Belloni AS, Vacca A, Roncali L, Nussdorfer
/372425a0. GG. Angiogenic activity of leptin in the chick embryo chorioal-
163. Martin SS, Qasim A, Reilly MP. Leptin resistance: a possible lantoic membrane is in part mediated by endogenous fibroblast
interface of inflammation and metabolism in obesity-related growth factor-2. Int J Mol Med. 2001;8(3):265–8.
402 Rev Endocr Metab Disord (2016) 17:389–403

180. Lago R, Gomez R, Lago F, Gomez-Reino J, Gualillo O. Leptin 198. Bhakoo HS, Milholland RJ, Lopez R, Karakousis C, Rosen F.
beyond body weight regulation—current concepts concerning its High incidence and characterization of glucocorticoid receptors
role in immune function and inflammation. Cell Immunol. in human malignant melanoma. J Natl Cancer Inst. 1981;66(1):
2008;252(1–2):139–45. doi:10.1016/j.cellimm.2007.09.004. 21–5.
181. Artwohl M, Roden M, Holzenbein T, Freudenthaler A, Waldhausl 199. Dobos J, Kenessey I, Timar J, Ladanyi A. Glucocorticoid receptor
W, Baumgartner-Parzer SM. Modulation by leptin of proliferation expression and antiproliferative effect of dexamethasone on hu-
and apoptosis in vascular endothelial cells. Int J Obes Relat Metab man melanoma cells. Pathol Oncol Res. 2011;17(3):729–34.
Disord. 2002;26(4):577–80. doi:10.1007/s12253-011-9377-8.
182. Ahima RS, Flier JS. Leptin. Annu Rev Physiol. 2000;62:413–37. 200. Spencer SJ, Tilbrook A. The glucocorticoid contribution to obesi-
doi:10.1146/annurev.physiol.62.1.413. t y. S t r e s s . 2 0 11 ; 1 4 ( 3 ) : 2 3 3 – 4 6 . d o i : 1 0 . 3 1 0 9
183. Grosfeld A, Andre J, Hauguel-De Mouzon S, Berra E, Pouyssegur /10253890.2010.534831.
J, Guerre-Millo M. Hypoxia-inducible factor 1 transactivates the 201. Livingstone DE, Jones GC, Smith K, Jamieson PM, Andrew R,
human leptin gene promoter. J Biol Chem. 2002;277(45):42953– Kenyon CJ, et al. Understanding the role of glucocorticoids in
7. doi:10.1074/jbc.M206775200. obesity: tissue-specific alterations of corticosterone metabolism
184. Brakenhielm E, Veitonmaki N, Cao R, Kihara S, Matsuzawa Y, in obese Zucker rats. Endocrinology. 2000;141(2):560–3.
Zhivotovsky B, et al. Adiponectin-induced antiangiogenesis and doi:10.1210/endo.141.2.7297.
antitumor activity involve caspase-mediated endothelial cell apo- 202. Shoag J, Haq R, Zhang M, Liu L, Rowe GC, Jiang A, et al. PGC-1
ptosis. Proc Natl Acad Sci U S A. 2004;101(8):2476–81. coactivators regulate MITF and the tanning response. Mol Cell.
185. Yokota T, Oritani K, Takahashi I, Ishikawa J, Matsuyama A, 2013;49(1):145–57. doi:10.1016/j.molcel.2012.10.027.
Ouchi N, et al. Adiponectin, a new member of the family of sol- 203. Price RA, Li WD, Zhao H. FTO gene SNPs associated with ex-
uble defense collagens, negatively regulates the growth of treme obesity in cases, controls and extremely discordant sister
myelomonocytic progenitors and the functions of macrophages. pairs. BMC Med Genet. 2008;9:4. doi:10.1186/1471-2350-9-4.
Blood. 2000;96(5):1723–32. 204. Ghosh S, Murinova L, Trnovec T, Loffredo CA, Washington K,
186. Kang JH, Lee YY, Yu BY, Yang BS, Cho KH, Yoon DK, et al. Mitra PS, et al. Biomarkers linking PCB exposure and obesity.
Adiponectin induces growth arrest and apoptosis of MDA-MB- Curr Pharm Biotechnol. 2014;15(11):1058–68.
231 breast cancer cell. Arch Pharm Res. 2005;28(11):1263–9. 205. De Coster S, van Larebeke N. Endocrine-disrupting chemicals:
187. Ogunwobi OO, Beales IL. Adiponectin stimulates proliferation associated disorders and mechanisms of action. J Environ Public
and cytokine secretion in colonic epithelial cells. Regul Pept. Health. 2012;2012:713696. doi:10.1155/2012/713696.
2006;134(2–3):105–13. doi:10.1016/j.regpep.2006.02.001. 206. Koch CA. EDITORIAL: BThe Koch’s^ view on the sense of taste
188. Bub JD, Miyazaki T, Iwamoto Y. Adiponectin as a growth inhib- in endocrinology. Rev Endocr Metab Disord. 2016;17(2):143–7.
itor in prostate cancer cells. Biochem Biophys Res Commun. doi:10.1007/s11154-016-9383-y.
2006;340(4):1158–66. doi:10.1016/j.bbrc.2005.12.103. 207. Brill MJ, Diepstraten J, van Rongen A, van Kralingen S, van
189. Arditi JD, Venihaki M, Karalis KP, Chrousos GP. Antiproliferative den Anker JN, Knibbe CA. Impact of obesity on drug metab-
effect of adiponectin on MCF7 breast cancer cells: a potential olism and elimination in adults and children. Clin
hormonal link between obesity and cancer. Horm Metab Res. Pharmacokinet. 2012;51(5):277–304. doi:10.2165/11599410-
2007;39(1):9–13. doi:10.1055/s-2007-956518. 000000000-00000.
190. Mantzoros CS, Trakatelli M, Gogas H, Dessypris N, Stratigos A, 208. James PA, Oparil S, Carter BL, Cushman WC, Dennison-
Chrousos GP, et al. Circulating adiponectin levels in relation to Himmelfarb C, Handler J, et al. evidence-based guideline for the
melanoma: a case–control study. Eur J Cancer. 2007;43(9):1430– management of high blood pressure in adults: report from the
6. doi:10.1016/j.ejca.2007.03.026. panel members appointed to the Eighth Joint National
191. Gallagher EJ, Fierz Y, Ferguson RD, LeRoith D. The pathway Committee (JNC 8). JAMA. 2014;311(5):507–20. doi:10.1001
from diabetes and obesity to cancer, on the route to targeted ther- /jama.2013.284427.
apy. Endocr Pract. 2010;16(5):864–73. doi:10.4158/EP10098. 209. McDonald E, Freedman DM, Alexander BH, Doody MM, Tucker
RA. MA, Linet MS, et al. Prescription diuretic use and risk of basal cell
192. Moore T, Carbajal S, Beltran L, Perkins SN, Yakar S, Leroith D, carcinoma in the nationwide U.S. radiologic technologists cohort.
et al. Reduced susceptibility to two-stage skin carcinogenesis in Cancer Epidemiol Biomarkers Prev. 2014;23(8):1539–45.
mice with low circulating insulin-like growth factor I levels. doi:10.1158/1055-9965.EPI-14-0251.
Cancer Res. 2008;68(10):2680–8. 210. Robinson SN, Zens MS, Perry AE, Spencer SK, Duell EJ, Karagas
193. Sadagurski MYS, Weingarten G, Holzenberger M, Rhodes CJ, MR. Photosensitizing agents and the risk of non-melanoma skin
Breitkreutz D, Leroith D, et al. Insulin-like growth factor 1 recep- cancer: a population-based case–control study. J Invest Dermatol.
tor signaling regulates skin development and inhibits skin 2013;133(8):1950–5. doi:10.1038/jid.2013.33.
keratinocyte differentiation. Mol Cell Biol. 2006;26(7):2675–87. 211. Kaae J, Boyd HA, Hansen AV, Wulf HC, Wohlfahrt J, Melbye M.
194. Chi M, Ye Y, Zhang XD, Chen J. Insulin induces drug resistance Photosensitizing medication use and risk of skin cancer. Cancer
in melanoma through activation of the PI3K/Akt pathway. Drug Epidemiol Biomarkers Prev. 2010;19(11):2942–9. doi:10.1158
Des Devel Ther. 2014;8:255–62. doi:10.2147/DDDT.S53568. /1055-9965.EPI-10-0652.
195. Kucera R, Treskova I, Vrzalova J, Svobodova S, Topolcan O, 212. Ruiter R, Visser LE, Eijgelsheim M, Rodenburg EM, Hofman A,
Fuchsova R, et al. Evaluation of IGF1 serum levels in malignant Coebergh JW, et al. High-ceiling diuretics are associated with an
melanoma and healthy subjects. Anticancer Res. 2014;34(9): increased risk of basal cell carcinoma in a population-based fol-
5217–20. low-up study. Eur J Cancer. 2010;46(13):2467–72. doi:10.1016/j.
196. Naspi A, Panasiti V, Abbate F, Roberti V, Devirgiliis V, Curzio M, ejca.2010.04.024.
et al. Insulin-like-growth-factor-binding-protein-3 (IGFBP-3) con- 213. Glatz M, Hofbauer GF. Phototoxic and photoallergic cutaneous
trasts melanoma progression in vitro and in vivo. PLoS One. drug reactions. Chem Immunol Allergy. 2012;97:167–79.
2014;9(6), e98641. doi:10.1371/journal.pone.0098641. doi:10.1159/000335630.
197. Jones SA, Morand EF. Glucocorticoids in 2015: new answers to 214. Moore DE. Drug-induced cutaneous photosensitivity: incidence,
old problems. Nat Rev Rheumatol. 2015. doi:10.1038 mechanism, prevention and management. Drug Saf. 2002;25(5):
/nrrheum.2015.176. 345–72.
Rev Endocr Metab Disord (2016) 17:389–403 403

215. Stern RS. Photocarcinogenicity of drugs. Toxicol Lett. 1998;102– line (DJM-1) in collagen gel matrix culture. J Invest Dermatol.
103:389–92. 2001;117(2):244–50. doi:10.1046/j.0022-202x.2001.01431.x.
216. Stern RS, Bigby M. An expanded profile of cutaneous reactions to 230. Rubin AI, Chen EH, Ratner D. Basal-cell carcinoma. N Engl J
nonsteroidal anti-inflammatory drugs. Reports to a specialty-based Med. 2005;353(21):2262–9. doi:10.1056/NEJMra044151.
system for spontaneous reporting of adverse reactions to drugs. 231. Marzuka AG, Book SE. Basal cell carcinoma: pathogenesis, epi-
JAMA. 1984;252(11):1433–7. demiology, clinical features, diagnosis, histopathology, and man-
217. Sanchez-Borges M, Capriles-Hulett A, Caballero-Fonseca F. Risk agement. Yale J Biol Med. 2015;88(2):167–79.
of skin reactions when using ibuprofen-based medicines. Expert 232. Espinosa P, Pfeiffer RM, Garcia-Casado Z, Requena C, Landi MT,
Opin Drug Saf. 2005;4(5):837–48. doi:10.1517 Kumar R, et al. Risk factors for keratinocyte skin cancer in patients
/14740338.4.5.837. diagnosed with melanoma, a large retrospective study. Eur J
218. Finch LE, Tomiyama AJ. Comfort eating, psychological stress, Cancer. 2015;53:115–24. doi:10.1016/j.ejca.2015.10.058.
and depressive symptoms in young adult women. Appetite. 233. Reinau D, Surber C, Jick SS, Meier CR. Epidemiology of basal
2015;95:239–44. doi:10.1016/j.appet.2015.07.017. cell carcinoma in the United Kingdom: incidence, lifestyle factors,
219. Bliss SA, Warnock JK. Psychiatric medications: adverse cutane- and comorbidities. Br J Cancer. 2014;111(1):203–6. doi:10.1038
ous drug reactions. Clin Dermatol. 2013;31(1):101–9. /bjc.2014.265.
doi:10.1016/j.clindermatol.2011.11.014. 234. Baxter JM, Patel AN, Varma S. Facial basal cell carcinoma. BMJ.
220. Borras L, Huguelet P. A case report of photosensitivity to 2012;345, e5342. doi:10.1136/bmj.e5342.
amisulpride. Prim Care Companion J Clin Psychiatry.
235. Cheng P, Weng SD, Chiang CH, Lai FJ. Relationship between
2007;9(2):153.
arsenic-containing drinking water and skin cancers in the
221. Warnock JK, Morris DW. Adverse cutaneous reactions to antipsy-
arseniasis endemic areas in Taiwan. J Dermatol. 2016;43(2):
chotics. Am J Clin Dermatol. 2002;3(9):629–36.
181–6.
222. Mennella JA, Bobowski NK, Reed DR. The development of sweet
236. Poschl G, Seitz HK. Alcohol and cancer. Alcohol Alcohol.
taste: from biology to hedonics. Rev Endocr Metab Disord.
2004;39(3):155–65.
2016;17(2):171–8. doi:10.1007/s11154-016-9360-5.
223. Iwasaki JK, Srivastava D, Moy RL, Lin HJ, Kouba DJ. The mo- 237. Saladi RN, Nektalova T, Fox JL. Induction of skin carcinogenicity
lecular genetics underlying basal cell carcinoma pathogenesis and by alcohol and ultraviolet light. Clin Exp Dermatol. 2010;35(1):7–
links to targeted therapeutics. J Am Acad Dermatol. 2012;66(5): 11. doi:10.1111/j.1365-2230.2009.03465.x.
e167–78. doi:10.1016/j.jaad.2010.06.054. 238. Song F, Qureshi AA, Gao X, Li T, Han J. Smoking and risk of skin
224. Bonilla X, Parmentier L, King B, Bezrukov F, Kaya G, Zoete V, cancer: a prospective analysis and a meta-analysis. Int J
et al. Genomic analysis identifies new drivers and progression Epidemiol. 2012;41(6):1694–705. doi:10.1093/ije/dys146.
pathways in skin basal cell carcinoma. Nat Genet. 2016;48(4): 239. Leonardi-Bee J, Ellison T, Bath-Hextall F. Smoking and the risk of
398–406. doi:10.1038/ng.3525. nonmelanoma skin cancer: systematic review and meta-analysis.
225. Rogers HW, Weinstock MA, Harris AR, Hinckley MR, Feldman Arch Dermatol. 2012;148(8):939–46. doi:10.1001
SR, Fleischer AB, et al. Incidence estimate of nonmelanoma skin /archdermatol.2012.1374.
cancer in the United States, 2006. Arch Dermatol. 2010;146(3): 240. Hughes MC, Olsen CM, Williams GM, Green AC. A prospective
283–7. doi:10.1001/archdermatol.2010.19. study of cigarette smoking and basal cell carcinoma. Arch
226. Makrantonaki EZC. Molecular mechanisms of skin aging: state of Dermatol Res. 2014;306(9):851–6. doi:10.1007/s00403-014-
the art. Acad Sci. 2007;1119:40–50. 1503-5.
227. Yan W, Wistuba II, Emmert-Buck MR, Erickson HS. Squamous 241. Henderson MT, Kubo JT, Desai M, David SP, Tindle H, Sinha
cell carcinoma—similarities and differences among anatomical AA, et al. Smoking behavior and association of melanoma and
sites. Am J Cancer Res. 2011;1(3):275–300. nonmelanoma skin cancer in the Women’s Health Initiative. J Am
228. Ibiebele TI, van der Pols JC, Hughes MC, Marks GC, Williams Acad Dermatol. 2015;72(1):190–1. doi:10.1016/j.
GM, Green AC. Dietary pattern in association with squamous cell jaad.2014.09.024. e3.
carcinoma of the skin: a prospective study. Am J Clin Nutr. 242. Trost SG, Owen N, Bauman AE, Sallis JF, Brown W. Correlates of
2007;85(5):1401–8. adults’ participation in physical activity: review and update. Med
229. Inoue T, Toda S, Narisawa Y, Sugihara H. Subcutaneous adipo- Sci Sports Exerc. 2002;34(12):1996–2001. doi:10.1249/01.
cytes promote the differentiation of squamous cell carcinoma cell MSS.0000038974.76900.92.
Reviews in Endocrine & Metabolic Disorders is a copyright of Springer, 2016. All Rights
Reserved.

Das könnte Ihnen auch gefallen