Review
Physiological cellular reprogramming and cancer

https://doi.org/10.1016/j.semcancer.2010.02.002Get rights and content

Abstract

The traditional approaches to cancer research and therapy have been primarily focused in the aspect of aberrant, uncontrolled, proliferation. Although this is clearly a very important issue, however, the emphasis on this characteristic has led to a relative neglect of an essential aspect of cancer biology: the alteration of normal differentiation processes. The oncogenic alterations that arise in an otherwise healthy cell lead to a whole reprogramming of the normal cellular fate and open a new pathologic developmental program. In this way cancer, reprogramming and cellular plasticity are tightly intertwined, since only some cells posses the necessary plasticity so as to allow the tumoral reprogramming to take place, and only some oncogenes have, in the right cellular context, the required tumoral reprogramming capacity. Research in the field of induced pluripotency is shedding a new light on the molecular mechanisms of tumor initiation and differentiation. In this review we discuss the latest findings in the area of cellular reprogramming and their implications from the point of view of tumor biology.

Introduction

The specification of cellular fate during development and differentiation is a dynamic and evolving process that starts with stem and progenitor cells and ends with terminal differentiation into a given specialized cellular type. Along this way there can be many cellular intermediates, some of them long-lived, and some very transient. In all cases, the maintenance of cellular identity is determined to a certain degree by the signals from the environment and, more importantly, in an intrinsic manner, by specific transcription factors that, together with epigenetic chromatin modifications, establish a defined gene expression profile and specific gene regulatory networks (GRNs).

Although many evidences about cellular plasticity had being accumulating for decades [1], [2], [3], [4], the latest findings in the field of reprogramming are finally shaking our beliefs about the stability of cellular identity and showing how switching to a different phenotype can be a lot easier than previously expected, and can have actual physiological relevance outside of the laboratory. Cancer is a clear case of pathological reprogramming where, from a normal tissue, a whole new differentiation branch is opened with its own hierarchy and structure [5], [6]. So, beyond aberrant deregulation of proliferation, tumoral reprogramming is an essential part of the tumorigenesis process, and it is closely dependent on the cellular plasticity of the cancer-initiating cells. Here we define cellular plasticity as the ability of cells (stem or differentiated) to adopt the biological properties (gene expression profile, phenotype, etc.) of other differentiated types of cells (belonging to the same or different lineages). This broad acception includes the property of competence, defined as the ability of stem cells and progenitors to give rise to their different descendant lineages during normal development. With this wide definition we intend to reflect the fact that the mechanisms that are important for stem cell competence during normal development are also at the basis of the plasticity of more differentiated types of cells, both in pathological processes and in experimentally induced fate-changing processes. In this article we will discuss the essential role of cellular plasticity in the origin and maintenance of cancerous cells. To this aim, we will first comment on the latest findings about normal developmental and experimentally induced plasticity, to consider afterwards how these properties connect with what we know about cancer biology.

Section snippets

Stem cell identity and lineage choice

Adult stem cells are in charge of replenishing the different types of specialized cells that compose the organism. Most of them fulfil this task during the entire life of the organism, thanks to their self-renewal capacity that allows them to divide asymmetrically originating a new identical multipotent stem cell and a multipotential progenitor that will give rise to all the variety of differentiated tissue cells. It is clear that there is a number of transcription factors that play a crucial

Cellular identity of differentiation intermediates

In some rare cases, adult stem cells are unipotent, and can only give rise to one type of differentiated cells (for example, spermatogonial stem cells, Fig. 1A). However, in most cases there are several types of specialized cells that can arise from a given stem cell type. So, once stem cells lose their self-renewal potential and enter the differentiation process by becoming multipotential progenitors, there are still many possibilities for them to follow. We have mentioned before that part of

Identity and plasticity of terminally differentiated cells

In general, the final cellular identity of any given differentiation pathway is stable and usually corresponds to a highly specialized cellular type with a particular physiological function. This actually seems logical since, from the point of view of physiology, it would not make sense that, under normal conditions, a specialized cell would have to be reprogrammed to give rise to a different cell type. So, in principle, plasticity, from the point of view of normal development, is a property

Transcriptional control of reprogramming in oncogenesis and induced pluripotency

Research in cancer has been a seminal source of knowledge and hypothesis for the fields of stem cell biology and developmental biology, including the fact that it was the study of teratocarcinomas what finally led to the identification of embryonic stem (ES) cells in blastocysts [31], [32], [33]. In the same way, there are several features of our current paradigm for understanding cancer development that have influenced or developed in parallel with the study of the reprogramming problem, and

Future prospects

The potential applications of pluripotent reprogramming to regenerative medicine are immense and tremendously varied. However, as we have highlighted in this review, the knowledge obtained in the research of the molecular and cellular mechanisms of reprogramming will also have deep implications for our understanding and treatment of cancer. Indeed, the two fields of research will continue to be closely intertwined and mutually dependent. As a way of an example, the main potential complication

Conflicts of interest

F.A.J. and R.J. have no relevant conflicts of interest to declare. C.C. was a scientific founder and owns a small percentage of stock of OncoStem Pharma, a small biotech company with interest in the cancer stem cell field.

Acknowledgments

We thank all members of labs 13 at IBMCC and B-15 at the Department of Physiology and Pharmacology for their helpful comments and constructive discussions. Research in the group is supported partially by FEDER (Fondo de Investigaciones Sanitarias PI080164), Proyectos Intramurales Especiales (CSIC), Fundación Mutua Madrileña and Junta de Castilla y León (SA060A09 and Proyecto Biomedicina 2009–2010). F.A.J. is the recipient of an FPU fellowship from Ministerio de Ciencia e Innovación.

References (107)

  • Y. Chen et al.

    The molecular mechanism governing the oncogenic potential of SOX2 in breast cancer

    J Biol Chem

    (2008)
  • Y. Tanaka et al.

    Forced expression of Nanog in hematopoietic stem cells results in a gammadeltaT-cell disorder

    Blood

    (2007)
  • R. Sridharan et al.

    Role of the murine reprogramming factors in the induction of pluripotency

    Cell

    (2009)
  • M. Wernig et al.

    c-Myc is dispensable for direct reprogramming of mouse fibroblasts

    Cell Stem Cell

    (2008)
  • J. Hanna et al.

    Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency

    Cell

    (2008)
  • S. Huang et al.

    Cancer attractors: a systems view of tumors from a gene network dynamics and developmental perspective

    Semin Cell Dev Biol

    (2009)
  • Y. Shi et al.

    A combined chemical and genetic approach for the generation of induced pluripotent stem cells

    Cell Stem Cell

    (2008)
  • Y. Zhao et al.

    Two supporting factors greatly improve the efficiency of human iPSC generation

    Cell Stem Cell

    (2008)
  • S. Alcantara Llaguno et al.

    Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model

    Cancer Cell

    (2009)
  • N.M. Joseph et al.

    The loss of Nf1 transiently promotes self-renewal but not tumorigenesis by neural crest stem cells

    Cancer Cell

    (2008)
  • H. Zheng et al.

    Induction of abnormal proliferation by nonmyelinating schwann cells triggers neurofibroma formation

    Cancer Cell

    (2008)
  • B.J. Huntly et al.

    MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors

    Cancer Cell

    (2004)
  • T.C. Somervaille et al.

    Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia

    Cancer Cell

    (2006)
  • C.W. So et al.

    MLL-GAS7 transforms multipotent hematopoietic progenitors and induces mixed lineage leukemias in mice

    Cancer Cell

    (2003)
  • F.C. Guibal et al.

    Identification of a myeloid committed progenitor as the cancer-initiating cell in acute promyelocytic leukemia

    Blood

    (2009)
  • D.J. Wong et al.

    Module map of stem cell genes guides creation of epithelial cancer stem cells

    Cell Stem Cell

    (2008)
  • C. le Viseur et al.

    In childhood acute lymphoblastic leukemia, blasts at different stages of immunophenotypic maturation have stem cell properties

    Cancer Cell

    (2008)
  • O. Bereshchenko et al.

    Hematopoietic stem cell expansion precedes the generation of committed myeloid leukemia-initiating cells in C/EBPalpha mutant AML

    Cancer Cell

    (2009)
  • S.M. Graham et al.

    Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro

    Blood

    (2002)
  • J.B. Kim et al.

    Oct4-induced pluripotency in adult neural stem cells

    Cell

    (2009)
  • V. Yechoor et al.

    Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes

    Dev Cell

    (2009)
  • X. Shen et al.

    Jumonji modulates polycomb activity and self-renewal versus differentiation of stem cells

    Cell

    (2009)
  • K. Hochedlinger et al.

    Nuclear reprogramming and pluripotency

    Nature

    (2006)
  • J.B. Gurdon et al.

    Nuclear reprogramming in cells

    Science

    (2008)
  • T. Graf et al.

    Forcing cells to change lineages

    Nature

    (2009)
  • C. Vicente-Duenas et al.

    The Role of cellular plasticity in cancer development

    Curr Med Chem

    (2009)
  • I. Sanchez-Garcia et al.

    The theoretical basis of cancer-stem-cell-based therapeutics of cancer: can it be put into practice?

    Bioessays

    (2007)
  • T. Reya et al.

    Stem cells, cancer, and cancer stem cells

    Nature

    (2001)
  • K.K. Niakan et al.

    Sox17 promotes differentiation in mouse embryonic stem cells by directly regulating extraembryonic gene expression and indirectly antagonizing self-renewal

    Genes Dev

    (2010)
  • H.H. Chang et al.

    Transcriptome-wide noise controls lineage choice in mammalian progenitor cells

    Nature

    (2008)
  • M. Hu et al.

    Multilineage gene expression precedes commitment in the hemopoietic system

    Genes Dev

    (1997)
  • T. Kalmar et al.

    Regulated fluctuations in nanog expression mediate cell fate decisions in embryonic stem cells

    PLoS Biol

    (2009)
  • C.H. Waddington

    The Strategy of the Genes

    (1957)
  • S. Huang

    Reprogramming cell fates: reconciling rarity with robustness

    Bioessays

    (2009)
  • M. Loose et al.

    Transcriptional networks regulating hematopoietic cell fate decisions

    Curr Opin Hematol

    (2007)
  • C.V. Laiosa et al.

    Determinants of lymphoid–myeloid lineage diversification

    Annu Rev Immunol

    (2006)
  • R. Ceredig et al.

    Models of haematopoiesis: seeing the wood for the trees

    Nat Rev Immunol

    (2009)
  • R. Briggs et al.

    Transplantation of living nuclei from blastula cells into enucleated frogs’ eggs

    Proc Natl Acad Sci USA

    (1952)
  • J.B. Gurdon

    The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles

    J Embryol Exp Morphol

    (1962)
  • S.L. Nutt et al.

    Commitment to the B-lymphoid lineage depends on the transcription factor Pax5

    Nature

    (1999)
  • Cited by (0)

    View full text