Review
Mouse models of Fanconi anemia

https://doi.org/10.1016/j.mrfmmm.2009.03.015Get rights and content

Abstract

Fanconi anemia is a rare inherited disease characterized by congenital anomalies, growth retardation, aplastic anemia and an increased risk of acute myeloid leukemia and squamous cell carcinomas. The disease is caused by mutation in genes encoding proteins required for the Fanconi anemia pathway, a response mechanism to replicative stress, including that caused by genotoxins that cause DNA interstrand crosslinks. Defects in the Fanconi anemia pathway lead to genomic instability and apoptosis of proliferating cells. To date, 13 complementation groups of Fanconi anemia were identified. Five of these genes have been deleted or mutated in the mouse, as well as a sixth key regulatory gene, to create mouse models of Fanconi anemia. This review summarizes the phenotype of each of the Fanconi anemia mouse models and highlights how genetic and interventional studies using the strains have yielded novel insight into therapeutic strategies for Fanconi anemia and into how the Fanconi anemia pathway protects against genomic instability.

Section snippets

Fanconi anemia

Fanconi anemia is a rare autosomal recessive disease with a complex spectrum of symptoms including congenital skeletal and renal anomalies, growth retardation, pigmentation abnormalities, fertility defects, aplastic anemia, and increased risk of acute myeloid leukemia and epithelial tumors (see “Fanconi Anemia and its Diagnosis” Auerbach, this issue). Progressive bone marrow failure and late-developing myeloid malignancies account for 90% of mortality in FA patients. Bone marrow failure in FA

Fanconi anemia pathway

The 13 FA proteins work as a complex signaling network that facilitates HR-mediated repair of DSBs caused by DNA ICLs and other types of replication stress (see “The Genetic and Molecular Basis of Fanconi Anemia” de Winter and Joenje, this issue, for more detail and a model). FANC A, B, C, E, F, G, L and M interact to form the FA core complex [19]. The FANCL subunit is an E3 ubiquitin ligase that monoubiquitylates FANCD2 and FANCI during S phase, particularly in response to genotoxic stress [48]

FancA−/− mice

Unlike FA patients, FancA−/− mice, created by deletion of exons 4–7, do not spontaneously display congenital anomalies or severe hematological abnormalities [12]. However, FancA−/− mice do have significantly reduced fertility due to hypogonadism [12]. Despite the mild phenotype, mouse embryonic fibroblasts (MEFs), derived from these mice are hypersensitive to mitomycin C (MMC) and accumulate large numbers of chromosomal aberrations in response to MMC [12], hallmark diagnostic criteria of FA

FancC−/− mice

Like the FancA−/− mice, genetic deletion of FancC in the mouse does not lead to skeletal abnormalities or spontaneous peripheral hematological abnormalities [11], [82]. However, FancC−/− mice are born with sub-Mendelian frequency and have a significantly increased incidence of microphthalmia, a congenital abnormality, if they are bred into a C57BL/6J background [6]. FancC−/− mice have impaired fertility [11], [82] due to impaired proliferation of germ cells during embryogenesis [51], similar to

FancG−/− mice

The phenotype of FancG−/− mice is virtually identical to that of FancA−/− and FancC−/− mice [18]. FancG−/− mice lack the characteristic congenital anomalies characteristic of FA and do not spontaneously develop hematological abnormalities or spontaneous cancer in the first year of life [37]. Like FancA−/− and FancC−/− mice, FancG−/− mice have reduced fertility due to impaired gametogenesis [37], [86], a characteristic of FA (see “Fanconi Anemia and its Diagnosis” Auerbach, this issue). Primary

FancD1/Brca2−/− mice

FANCD1 is identical to BRCA2 [34]. FANCD1/BRCA2 interacts with RAD51 [45] and is required for HR-mediated repair of DNA DSBs [49], [78]. In humans, hapoloinsufficiency of BRCA2 leads to a dramatically increased risk of breast, ovarian and pancreatic cancer. Genetic deletion of FancD1/Brca2 in the mouse results in embryonic lethality [70]. Homozygous deletion of exon 27 of FancD1/Brca2 prevents the interaction of FANCD1/BRCA2 with FANCD2 [2]. Hematopoietic cell function is significantly

FancD2−/− mice

FancD2−/− mice are viable [32], indicating that FANCD2 is not required for mammalian development. FancD2−/− mice are born with sub-Mendelian frequency and display pre- and post-natal growth retardation. The severity of the phenotype of the mice is dependent upon their genetic background, with a more severe phenotype emerging in C57BL/6J than in 129S4, indicating the existence of modifying loci. Like FancA−/−, FancC−/− and FancG−/− mice and FA patients, cells from FancD2−/− mice are

Usp1−/− mice

The deubiquitylating enzyme, USP1 (ubiquitin-specific protease 1), was recently demonstrated to regulate the level of monoubiquitylated FANCD2 and FANCI proteins [55], [76]. Inhibition of Usp1 by siRNA knockdown in human cell lines leads to an accumulation of monoubiquitylated isoforms of FANCD2 and FANCI proteins. Unexpectedly, disruption of Usp1 in chicken DT40 cells results in hypersensitivity to DNA interstrand crosslinking agents, similar to that observed in cells with mutations in FA

Genetic studies: double mutant mice

FancA−/−;FancC−/− mice and cells derived from them are phenotypically identical to single mutants [56]. This provides crucial genetic evidence that FANCA and FANCC are epistatic. FancC−/− mice were crossed into a Sod1−/− background to delete Cu/Zn superoxide dismutase and increase endogenous oxidative stress [25]. The double mutant mice display bone marrow hypocellularity due to a loss of committed progenitor cells, resulting in anemia and leucopenia. FancC−/−;Sod1−/− bone marrow progenitor

Interventional studies to elucidate the function of the FA pathway

FancC−/− mice chronically exposed to a sublethal dose of the crosslinking agent MMC develop progressive pancytopenia due to bone marrow failure [8], identical to the spontaneous symptoms of FA. MMC exposure depletes the bone marrow of CD34+ cells, but not CD34 [7], suggesting that early hematopoietic progenitors are particularly vulnerable to crosslink damage. FancC−/− mice chronically treated with MMC offer a good model in which to study therapeutic interventions for FA. Transplantation of

Utilizing the mouse models to discover therapeutic options for Fanconi anemia

Hematological disease and malignancies are the most common cause of death in FA (see “Fanconi Anemia and its Diagnosis” Auerbach, this issue). This can be prevented by bone marrow transplantation, indicating that FA is a prime candidate disease for gene therapy (see “Finding the Needle in the Hay Stack: Hematopoietic Stem Cells in Fanconi Anemia”, Muller and Williams, this issue, for more information on gene therapy in FA). Indeed, retroviral transduction of FancA−/− hematopoietic stem cells

Summary

In conclusion, FA mouse models now exist, resulting from targeted disruption of the FancA, FancC, FancG, FancD1, FancD2, or Usp1 gene. The mouse models exhibit some but not all of the developmental and hematologic manifestations of human FA patients (Table 1). While FA patients develop spontaneous hematologic failure, most FA mouse models have relatively normal hematologic function, though anemia can be elicited by in vivo exposure to crosslinking agents. Mouse models will be especially useful

Conflict of interest

The authors declare that there are no conflicts of interest.

Acknowledgments

L.J.N. is supported by The Ellison Medical Foundation (AG-NS-0303-05) and NIEHS (R01 ES016114). K.P. and A.D.D. were supported by NIH grant U19AI067751.

References (91)

  • I. Garcia-Higuera et al.

    Interaction of the Fanconi anemia proteins and BRCA1 in a common pathway

    Mol. Cell

    (2001)
  • B.C. Godthelp et al.

    Inducibility of nuclear Rad51 foci after DNA damage distinguishes all Fanconi anemia complementation groups from D1/BRCA2

    Mutat. Res.

    (2006)
  • K.A. Gush et al.

    Phenotypic correction of Fanconi anemia group C knockout mice

    Blood

    (2000)
  • S. Hadjur et al.

    Increased sensitivity of Fancc-deficient hematopoietic cells to nitric oxide and evidence that this species mediates growth inhibition by cytokines

    Blood

    (2003)
  • S. Hadjur et al.

    Defective hematopoiesis and hepatic steatosis in mice with combined deficiencies of the genes encoding Fancc and Cu/Zn superoxide dismutase

    Blood

    (2001)
  • L.S. Haneline et al.

    Multiple inhibitory cytokines induce deregulated progenitor growth and apoptosis in hematopoietic cells from Fac−/− mice

    Blood

    (1998)
  • L.S. Haneline et al.

    Loss of FancC function results in decreased hematopoietic stem cell repopulating ability

    Blood

    (1999)
  • L.S. Haneline et al.

    Retroviral-mediated expression of recombinant Fancc enhances the repopulating ability of Fancc−/− hematopoietic stem cells and decreases the risk of clonal evolution

    Blood

    (2003)
  • N. Hogg et al.

    Nitric oxide and lipid peroxidation

    Biochim. Biophys. Acta

    (1999)
  • J.M. Kim et al.

    Inactivation of murine Usp1 results in genomic instability and a fanconi anemia phenotype

    Dev. Cell

    (2009)
  • X. Li et al.

    Ex vivo culture of Fancc−/− stem/progenitor cells predisposes cells to undergo apoptosis, and surviving stem/progenitor cells display cytogenetic abnormalities and an increased risk of malignancy

    Blood

    (2005)
  • X. Li et al.

    Fanconi anemia type C-deficient hematopoietic stem/progenitor cells exhibit aberrant cell cycle control

    Blood

    (2003)
  • X. Li et al.

    Continuous in vivo infusion of interferon-gamma (IFN-gamma) preferentially reduces myeloid progenitor numbers and enhances engraftment of syngeneic wild-type cells in Fancc−/− mice

    Blood

    (2004)
  • Y. Li et al.

    Mesenchymal stem/progenitor cells promote the reconstitution of exogenous hematopoietic stem cells in Fancg−/− mice in vivo

    Blood

    (2009)
  • J. Maciejewski et al.

    Fas antigen expression on CD34+ human marrow cells is induced by interferon gamma and tumor necrosis factor alpha and potentiates cytokine-mediated hematopoietic suppression in vitro

    Blood

    (1995)
  • M.E. Moynahan et al.

    BRCA2 is required for homology-directed repair of chromosomal breaks

    Mol. Cell

    (2001)
  • L.U. Muller et al.

    Rapid lentiviral transduction preserves the engraftment potential of Fanca(−/−) hematopoietic stem cells

    Mol. Ther.

    (2008)
  • S. Navarro et al.

    Hematopoietic dysfunction in a mouse model for Fanconi anemia group D1

    Mol. Ther.

    (2006)
  • L.J. Niedernhofer et al.

    Malondialdehyde, a product of lipid peroxidation, is mutagenic in human cells

    J. Biol. Chem.

    (2003)
  • S.M. Nijman et al.

    The deubiquitinating enzyme USP1 regulates the Fanconi anemia pathway

    Mol. Cell

    (2005)
  • M. Noll et al.

    Fanconi anemia groups A and C double-mutant mice: functional evidence for a multi-protein Fanconi anemia complex

    Exp. Hematol.

    (2002)
  • V.H. Oestergaard et al.

    Deubiquitination of FANCD2 is required for DNA crosslink repair

    Mol. Cell

    (2007)
  • R.K. Rathbun et al.

    Inactivation of the Fanconi anemia group C gene augments interferon-gamma-induced apoptotic responses in hematopoietic cells

    Blood

    (1997)
  • P. Rio et al.

    In vivo proliferation advantage of genetically corrected hematopoietic stem cells in a mouse model of Fanconi anemia FA-D1

    Blood

    (2008)
  • P. Rio et al.

    In vitro phenotypic correction of hematopoietic progenitors from Fanconi anemia group A knockout mice

    Blood

    (2002)
  • F. Rosselli et al.

    Abnormal lymphokine production: a novel feature of the genetic disease Fanconi anemia. II. In vitro and in vivo spontaneous overproduction of tumor necrosis factor alpha

    Blood

    (1994)
  • Y. Si et al.

    Continuous in vivo infusion of interferon-gamma (IFN-gamma) enhances engraftment of syngeneic wild-type cells in Fanca−/− and Fancg−/− mice

    Blood

    (2006)
  • A.M. Skinner et al.

    CXCR4 induction in hematopoietic progenitor cells from Fanca(−/−), -c(−/−), and -d2(−/−) mice

    Exp. Hematol.

    (2008)
  • A. Smogorzewska et al.

    Identification of the FANCI protein, a monoubiquitinated FANCD2 paralog required for DNA repair

    Cell

    (2007)
  • M.A. Whitney et al.

    Germ cell defects and hematopoietic hypersensitivity to gamma-interferon in mice with a targeted disruption of the Fanconi anemia C gene

    Blood

    (1996)
  • J.C. Wong et al.

    Cloning and analysis of the mouse Fanconi anemia group A cDNA and an overlapping penta zinc finger cDNA

    Genomics

    (2000)
  • K. Yamada et al.

    Phenotype correction of Fanconi anemia group A hematopoietic stem cells using lentiviral vector

    Mol. Ther.

    (2003)
  • Y. Yang et al.

    Targeted disruption of the murine Fanconi anemia gene, Fancg/Xrcc9

    Blood

    (2001)
  • X. Zhang et al.

    Hypoxia-reoxygenation induces premature senescence in FA bone marrow hematopoietic cells

    Blood

    (2005)
  • X. Zhang et al.

    The Fanconi anemia proteins functionally interact with the protein kinase regulated by RNA (PKR)

    J. Biol. Chem.

    (2004)
  • Cited by (127)

    • A Fanca knockout mouse model reveals novel Fancd2 function

      2024, Biochemical and Biophysical Research Communications
    View all citing articles on Scopus
    View full text