Review
Article Series: Cancer and Metabolism
PTEN in cancer, metabolism, and aging

https://doi.org/10.1016/j.tem.2012.11.002Get rights and content

Recent reports on mice with systemic overexpression of the tumor-suppressor PTEN (phosphatase and tensin homolog) have expanded our understanding of its physiological functions. Pten transgenic mice present increased energy expenditure, decreased adiposity, improved insulin sensitivity upon high-fat feeding or with aging, and extended lifespan. This has led to new mechanistic insights about the role of PTEN in metabolism. Interestingly, PTEN promotes oxidative phosphorylation and decreases glycolysis, thus preventing the metabolic reprogramming characteristic of cancer cells, which might be relevant to PTEN-mediated cancer protection. PTEN also upregulates UCP1 expression in brown adipocytes, which enhances their nutrient burning capacity and decreases adiposity and associated pathologies. The newly discovered effects of PTEN on metabolism open new avenues for exploration relevant to cancer, obesity, diabetes, and aging.

Section snippets

PTEN and cancer protection

PTEN is a well-established tumor-suppressor gene, being one of the most frequently mutated genes in human tumors and with germline mutations causing cancer-predisposition syndromes 1, 2, 3. The protein is expressed in all tissues in the body and contains a tensin-like domain and a phosphatase catalytic domain [4]. During tumor development, PTEN mutations and deletions often occur that inactivate or decrease PTEN function [3]. Genetic inactivation of PTEN is frequently found in glioblastomas,

PTEN and obesity

Overexpression of PTEN in mice results in reduced body weight 12, 13. This phenotype is not surprising considering that PTEN antagonizes a main anabolic pathway, such as the insulin-activated PI3K/AKT pathway. Strikingly, however, PTEN overexpression results in mice that are hyperphagic, which, combined with their reduced body weight, suggests a poorer capacity to store the energy of nutrients and thus lower metabolic efficiency. Indeed, calorimetric measurements demonstrated an increased

PTEN and longevity

Given the aforementioned phenotypes associated with increased Pten dosage, namely protection from cancer and obesity, PTEN might also have an effect on longevity. Indeed, Ptentg mice exhibit higher median and maximal lifespans, a phenomenon observed both in males and females [12]. The effect of PTEN on longevity is, however, independent of its action as a tumor suppressor, because when longevity is measured only in those mice that do not develop cancer (longevity of cancer-free mice), Pten

Insulin signaling versus long-term insulin sensitivity

The fact that many mouse models with reduced insulin signaling, including Ptentg mice, have increased lifespan, begs the question of why these mice do not develop life-limiting pathologies associated with insulin resistance. This conundrum can be explained by the existence of negative feedback loops within the IIS pathway, which are most relevant under conditions of chronic nutrient overload, or during aging. In particular, negative feedback loops emanate from FOXO and S6K that inhibit the

The ‘anti-Warburg effect’ of PTEN

Cancer cells, in addition to deregulated control of cell proliferation and survival, must also reprogram their metabolism to fuel cell growth and division [29]. The most remarkable metabolic change of cancer cells is the so-called ‘Warburg effect’, by which cells catabolize large amounts of glucose through glycolysis. This massive increase in glycolytic flux maintains the necessary ATP levels and, at the same time, promotes the generation of anabolic intermediates [30]. The direct connection

Specific effects of PTEN on brown adipose tissue (BAT)

In the context of studying glucose uptake in vivo, it was found that the BAT of Ptentg mice was abnormally active compared to wt mice (Figure 2) [12]. Brown adipocytes constitute the main cell type of the BAT and are also found in clusters interspersed within the white adipose tissue (WAT) 40, 41. The main function of brown adipocytes is to burn nutrients either in response to cold (cold-induced thermogenesis) or in response to nutritional excess (diet-induced thermogenesis) 40, 41. Uncoupling

Concluding remarks

The new metabolic roles of PTEN, as a negative regulator of glycolysis and a positive regulator of energy expenditure, are part of a general trend of discoveries by which oncogenes and tumor suppressors are found to be intimately connected to metabolism. This should not be surprising, given the fact that cell growth and proliferation are tightly dependent on metabolism. A major pending question for the immediate future is to dissect the extent to which the metabolic effects of tumor suppressors

Acknowledgments

Work in the laboratory of M.S. is funded by the Spanish National Cancer Research Centre (CNIO) and by grants from the Spanish Ministry of Economy (SAF), the European Commission (European Research Council Advanced Grant), the Regional Government of Madrid, the Botin Foundation, the Ramon Areces Foundation, and the AXA Foundation.

References (51)

  • R.J. DeBerardinis

    The biology of cancer: metabolic reprogramming fuels cell growth and proliferation

    Cell Metab.

    (2008)
  • K. Bensaad

    TIGAR, a p53-inducible regulator of glycolysis and apoptosis

    Cell

    (2006)
  • M.S. Song

    Nuclear PTEN regulates the APC-CDH1 tumor-suppressive complex in a phosphatase-independent manner

    Cell

    (2011)
  • A. Frontini et al.

    Distribution and development of brown adipocytes in the murine and human adipose organ

    Cell Metab.

    (2010)
  • J. Nedergaard et al.

    The changed metabolic world with human brown adipose tissue: therapeutic visions

    Cell Metab.

    (2010)
  • M.P. Mattson

    Perspective: does brown fat protect against diseases of aging?

    Ageing Res. Rev.

    (2010)
  • L.C. Cantley et al.

    New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway

    Proc. Natl. Acad. Sci. U.S.A.

    (1999)
  • M.C. Hollander

    PTEN loss in the continuum of common cancers, rare syndromes and mouse models

    Nat. Rev. Cancer

    (2011)
  • M.S. Song

    The functions and regulation of the PTEN tumour suppressor

    Nat. Rev. Mol. Cell Biol.

    (2012)
  • A. Alimonti

    Subtle variations in Pten dose determine cancer susceptibility

    Nat. Genet.

    (2010)
  • N. Chalhoub et al.

    PTEN and the PI3-kinase pathway in cancer

    Annu. Rev. Pathol.

    (2009)
  • R. Zoncu

    mTOR: from growth signal integration to cancer, diabetes and ageing

    Nat. Rev. Mol. Cell Biol.

    (2011)
  • P.L. de Keizer

    Forkhead box O as a sensor, mediator, and regulator of redox signaling

    Antioxid. Redox Signal.

    (2011)
  • A. Pal

    PTEN mutations as a cause of constitutive insulin sensitivity and obesity

    N. Engl. J. Med.

    (2012)
  • T.R. Castaneda

    Metabolic control by S6 kinases depends on dietary lipids

    PLoS ONE

    (2012)
  • Cited by (165)

    View all citing articles on Scopus
    View full text