Introduction
Fanconi anemia (FA) is an autosomal recessive human disorder that was first described by Guido Fanconi and is characterized by aplastic anemia, skeletal defects, growth retardation and a predisposition to cancer [
1]. FA arises due to a mutation in genes involved in FA repair [
2]. The pathway resolves interstrand crosslinks (ICLs), and is regulated by 23 complementation groups of which eight proteins (FANC-A, B, C, E, F, G, L, M) in association with other FA-associated proteins (FAAPs), forms a ubiquitin E3 ligase complex called the FA core complex [
3,
4,
5]. This complex assembles at the site of the ICL and monoubiquitinates the FANCI-D2 complex [
4,
6]. Thereafter, a cascade of proteins is recruited to resolve the ICL in an error-free manner [
7].
Human
FANCG is localized to chromosome 9 and was identified to be identical to
XRCC9, which complemented the MMC-sensitive Chinese hamster mutant UV40 [
8,
9,
10]. Murine FANCG protein is 83% similar to its human counterpart and its expression in human
Fancg-deficient lymphoblasts corrects their sensitivity to crosslinks [
11]. The protein predominantly localizes to the nucleus and has two leucine zipper motifs [
12]. The protein is most expressed in the testis, spleen and thymus [
11]. FANCG interacts with FANCA, FANCC [
13,
14], is required for the assembly of the FA core complex [
15,
16] and HR-mediated repair of specific double strand breaks (DSBs) [
17].
Genetically engineered mouse models (GEMMs) are an invaluable source to study human diseases as well as uncover the physiological relevance of genes and their regulatory regions in a systemic setting [
18,
19]. FA is phenotypically heterogenous and mouse models have since been created to recapitulate the FA patient phenotype and understand the relevance of each FA gene in maintaining tissue homeostasis [
3,
20,
21,
22]. These GEMMs also provide a rich source to establish primary as well as transformed cell lines enabling an in-depth
in vitro characterization of FA proteins and their interactome as part of the DNA damage response (DDR) network. To date, all FA mouse models display reduced fertility and cell line-based hypersensitivity to ICLs [
3,
20]. Additionally, most models also demonstrate a partial embryonic lethal phenotype, but severe anemia, bone marrow failure and risk of cancer, as observed in FA patients, is not recapitulated in mice, which likely relates to differences in metabolism, life expectancy, and use of alternative ICL repair pathways [
20]. Nevertheless, these GEMMs provide a solid platform to screen and test for new therapies, including immunotherapy, that have the potential to improve current standard treatments.
The improvements in genome editing via the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) protein 9 system over the past decade have been phenomenal [
23,
24]. Borrowed from the prokaryotic cells, the system consists of one or more sequence-specific guide RNAs (gRNA) that identify the target DNA sequence using protospacers adjacent motifs (PAMs) and guide the Cas9 nuclease to cleave this target sequence [
25,
26,
27]. This results in DSBs that are subsequently repaired, introducing the required genetic modification. CRISPR-Cas9 is highly specific, easy to engineer and a multiplexable tool thereby reducing the time and cost of making models, especially animal models [
28,
29].
Former
Fancg-deficient mouse models have been generated on mixed backgrounds, using selection cassettes and partial disruption of the
Fancg locus [
30,
31], presenting genotype-phenotype disparities up for discussion. Furthermore, given that immunotherapy is on the forefront of new treatment modalities in genetically instable cancers, the genetic background of these GEMMs becomes relevant in excluding genetic confounders. To accomplish this goal, we generated a C57BL/6J
Fancg-KO (KO) mouse model using CRISPR/Cas9 technology that employed two RNA guides that induced targeted double strand breaks (DSBs) at each end of the
Fancg locus. The GEMM was generated and maintained on an immunologically well-defined C57BL/6J background to exclude any confounding phenotypes related to mixed genetic backgrounds. Successful CRISPR/Cas9 mediated ablation of the
Fancg locus was confirmed by DNA genotyping and cDNA expression. Homozygous mutants were born sub-Mendelian, indicative of embryonic lethality and demonstrated infertility, hematopoietic defects and skewing of hematopoiesis towards the myelopoietic compartment.
Fancg-KO PreB cells and mouse embryonic fibroblasts (MEFs) displayed hypersensitivity to crosslinking agents-cisplatin and mitomycin C. KO MEFs also displayed levels of γ-H2AX upon cisplatin exposure. Finally, reconstitution with a flagged
Fancg cDNA- construct fully rescued sensitivity of KO cells to cisplatin, further validating the C57BL/6J
Fancg-KO mouse model for further fundamental and applied studies in FA research.
Discussion
Crosslinking agents are widely applied in cancer therapy as they introduce one of the most toxic DNA lesions i.e., interstrand crosslinks (ICLs). These lesions are effectively repaired by the Fanconi anemia (FA) repair pathway. Inactivation of one of the FA repair components can lead to pleiotropic defects such as aplastic anemia, congenital abnormalities and skeletal retardation in FA patients. While genetically engineered mouse models (GEMMs) fail to recap all of the phenotypes seen in FA patients, the common hypersensitivity to ICLs studies closely recapitulate those of FA patients and cells. In this regard, FA GEMMs and cell lines thereof provide ideal model systems to study the relevance of FA-ICL repair in stem cell maintenance and hematopoiesis. Applying the CRISPR/Cas9 technology in C57BL/6J zygotes we generated and characterized a novel, genetically and immunologically well-defined C57BL/6J
Fancg-KO mouse model. To exclude the possibility of generating hypomorphic gene products, the entire
Fancg locus was deleted. Ablation of the
Fancg locus was verified by genotyping and RNA expression. In contrast to conventional KO approaches, this gene targeting strategy excludes experimental confounders related to mixed backgrounds and the co-segregating of modulators and other unrelated genetically linked phenotypesx [
30,
31]. The exclusion of such confounder warrants conclusive genotype-phenotype interactions.
To minimize genetic drifting,
Fancg mutant mice were maintained on a C57BL/6J background. To derive homozygous mutants, heterozygous
Fancg mice were intercrossed. KO mice were obtained at a sub-Mendelian frequency, suggesting that homozygous condition causes embryonic lethality, similar to recent observations made for a
Fanci-/- model [
21] (
Figure 1E), but contrasting previous observations made in independent
Fancg mouse models [
30,
31]. In these reports,
Fancg-/- mice were born at a normal Mendelian frequency. However, these studies used mice with mixed backgrounds [
30,
31], a potential confounder that could mask specific genotype-phenotype interactions. As we observe expected
Fancg-KO numbers at E14.5 but not at weaning (E21), the FANCG-deficient embryos apparently experience a specific stress condition prior to weaning, a phenotype that necessitates future in depth studies. Nevertheless, consistent with other FA mouse models in previous studies, our KO mice were also infertile with gross germ cell defects and reduced size of testes/ovaries which might be due to the RAC1-based exhaustion of primordial germ cells in the absence of FANCG [
43]. Otherwise
Fancg-KO developed normally without any overt macroscopic abnormalities. Consistently, none of the tissues examined microscopically displayed any obvious pathological features.
Regarding the involvement of FA pathway in hematopoiesis, the
Fancg-/- mouse models studied thus far did not reveal hematological defects based on blood cell counts, hemoglobin analysis and blood smears [
30,
31,
36,
44,
45]. Similar to other FA mice [
22,
38,
46,
47], the blood smears of our KO mice showed no signs of anemia indicating that unlike FA patients spontaneous anemia inflicted by reactive crosslinking metabolites does not occur in mice [
40]. Therefore, the anemic phenotype in humans might be visualized better by the disruption of more than one gene in the FA pathway in mice [
36,
48], as shown previously in compound GEMMs lacking
Fancc and
Fancg [
36].
Regarding hematopoietic subsets, the bone marrow of our KO mice shows a significant reduction in early progenitors i.e., LSK subset which is in line with observations made by Barroca
et al. in their
Fancg-/- mice [
44] and other FA mouse models [
48,
49,
50]. We also noted similarities regarding the common lymphoid and myeloid progenitors (CLP & CMP), although we did not observe a significant reduction in the GMP and MEP subsets. While Barroca
et al. use CD34 and CD135 (
Flk2) to define their short term and long-term HSCs [
44], we utilized a more comprehensive panel including SLAM receptors [
51] CD150 and CD48 to define our HSC and MPP populations as described by Wilson
et al. [
35]. Interestingly, their short-term HSCs (LSKCD34
+Flk2
-) are equivalent to our MPP1-3 and their MPPs (LSKCD34
+FLk2
+) equivalent to our MPP4 populations, and display similar behaviour. However, unlike their long-term HSCs (LSKCD34
-Flk2
-), we do not see a significant drop in our HSCs (LSKCD34
-CD48
-CD150
+CD135
-). In summary, both models revealed an important role of FANCG in myelopoiesis and lymphopoiesis. Additionally, our data demonstrates that lymphoid progenitors (CLPs and MPP4s) are compromised, likely to sustain the differentiation of myeloid progenitors GMP and MEPs, a hallmark of stressed hematopoiesis.
The most stable FA phenotype relates to the hypersensitivity to crosslinking agents [
21]. PreBs and MEFs derived from our KO mice were, as expected, hypersensitive to both cisplatin and MMC. We were able to rescue this sensitivity by retroviral transduction of a wild type murine
Fancg cDNA. In addition, this construct possesses Flag tags flanking FANCG which can be utilized to perform immunoprecipitation-based studies and provides a solution for the lack of efficient anti-mFANCG antibodies in the market for biochemistry and molecular biology-based experiments.
In summary, we present here a new, genetically and immunologically well-defined C57Bl/6J Fancg-KO mouse model carrying a complete ablation of the Fancg locus that presents key characteristics of FA phenotypes. In addition to the mouse model, cells derived thereof as well as their reconstituted counterparts provide an opportunity to study FA repair, particularly the role of FANCG under specific ICL stress conditions caused by metabolites as well as chemotherapeutics.
Materials and Methods
Generation of Fancg-/- Mouse
A
Fancg-KO mouse model was generated wherein C57BL/6J zygotes were injected with
in vitro transcribed Cas9 mRNA and two guide RNAs (gRNAs) targeting
Fancg [
29,
52]. The 2 gRNAs targeting exon 1 and 14 were designed using the crispr.mit.edu tool and transcribed
in vitro from PCR templates that had a T7 promoter. To distinguish wildtype and homozygous mutants, we established a three primer PCR strategy. Pups born were selected for the knock out allele, backcrossed once onto C57BL6/J and maintained for the desired experimental genotype.
Generation of Primary Cell-Lines and Cell Culture
Timed-matings of
Fancg+/- females were set up with corresponding males to obtain embryos. On day 14.5 of gestation, embryos were isolated to prepare primary mouse embryonic fibroblasts (pMEFs). From Fetal liver, Pre-B cell cultures were established by culturing on irradiated ST2 feeder cells in complete IMDM medium (Iscoves, supplemented with 8% fetal calf serum (FCS), 50 μM 2-mercapthoethanol, penicillin/streptomycin) supplemented with IL-7 containing supernatant [
53]. Primary MEFs (2, from independent embryos per genotype) were isolated using Trypsin and cell strainers and cultured under low (3%) oxygen condition, with 5% CO2 at 37°C. To immortalize MEFs, pMEFs were transduced with a lentivirus encoding a p53-specific shRNA [
54]. The immortalized
Trp53kd MEFs were grown in complete IMDM medium under normal oxygen levels with 5% CO2 at 37°C.
PCR Genotyping
Genomic DNA was extracted from mouse toe tips and used as template for polymerase chain reaction (PCR) genotyping. The PCR protocol was: 95°C for 2 min and then 40 cycles of 95°C for 30 s, 58°C for 40 s and 72°C for 1 min; last 5 min at 72°C.
qRT-PCR
RNeasy mini (Qiagen) was used to isolate total RNA from wild type and
Fancg-KO MEFs for qRT-PCR. The cDNA libraries were synthesized using Invitrogen Superscript III kit and random hexamer primers.
Fancg was amplified with high fidelity PfuUltra Hotstart DNA polymerase (Stratagene) using gene specific primers (id: 253735709c1) from PrimerBank database [
55].
Immunofluorescence of Bone Marrow (BM) Cells
Mice were euthanized at indicated age (8-16 weeks), and BM cells from femur and tibia were flushed out using 21 Gauge syringes in cold PBEA buffer (1x PBS, 0.5% BSA, 2mM EDTA, 0.02% Sodium Azide). Eythrocytes were eliminated from the BM cells using an erylysis buffer (NH4Cl, KHCO3, 0.5M EDTA) and passed through a 70 µm filter. 10 million cells were used for staining. The following antibodies were used: Mouse Lineage Cell Detection Cocktail biotin antibody (1:40) followed by c-kit-APC (Clone 2B8, eBioscience), Streptavidin-APC-Cy7 (Southern Biotech), CD135-PE (Clone A2F10), CD48-PE-Dazzle (Clone HM48-1), Sca1-PE-Cy7 (Clone D7), CD34-FITC (Clone RAM34, Invitrogen, 1:100), CD127-BV421 (Clone A7R34), CD150-BV650 (Clone TC15-12F12.2), CD16/32-BV786 (clone 2.4G2, BD Bioscience). Dead cells were excluded by 7AAD staining. All the antibodies for FACS analysis were from Biolegend and used 1:200, unless otherwise specified.
Histopathology
Spleen, thymus, sternum and testis/ovary were collected and fixed in ethanol-(not glacial) acetic acid-formalin (EAF). Samples were embedded in paraffin, sections of 4 µm thickness were made and stained for Hematoxylin and eosin (H&E). The sections were reviewed with a Zeiss AxiosKOp2 Plus microscope (Carl Zeiss Microscopy, Jena, Germany), and images were captured with a Zeiss AxioCam HRc digital camera and processed with AxioVision 4 software (both from Carl Zeiss Vision, München, Germany). The scale bars were set at 50 μm. Blood smears were stained with Wright-Giemsa stain.
PreB Survival Assay
Survival assay was performed as described previously [
53]. Briefly, 10
5 pre-B cells were seeded in 12-wells plates containing an ST2 feeder layer and 1 ml complete medium and IL-7 in the continuous presence of different doses of the cisplatin or MMC. To determine cell survival, cells were harvested after 3 days of culture and stained with DAPI. The number of DAPI-negative cells was measured on a Fortessa (Becton Dickinson). Data analysis was performed with FlowJo software.
IncuCyte Proliferation Assay
The IncuCyte ZOOM instrument (Essen Bioscience) live cell imaging system was used to monitor cell growth of MEFs. 250 cells were plated in a 96 Greiner micro clear black well plate and imaged every 4 hours (hrs) in the presence or absence of cisplatin or MMC. The default software parameters for a 96 well plate with a 10x objective were used for imaging. The IncuCyte software was used to calculate mean confluence from four non-overlapping bright phase images of each well.
γ-H2AX Immunofluorescence
Immunofluorescence was performed as described previously. Cells were grown on coverslips for a day before the treatment with 20 µM CsPt for 1 hr. After treatment, one batch was placed on ice and the second batch was refreshed with medium. After 4 hrs, both batches of cells were washed with PBS and pre-extracted with PBS/0.5%TritonX-100) on ice for 1 minute. Cells were then fixed using 4% Formaldehyde for 15 mins at room temperature (RT). Fixed cells were then incubated with primary Abs against γ-H2AX at RT for 90 mins. Cells were washed and incubated with secondary Abs alongside DAPI for 1 hr at RT. After washing, coverslips were mounted onto glass slides using Aqua Poly/Mount. Pictures were taken with a Zeiss AxioObserver Z1 inverted microscope using a 63x lens equipped with a cooled Hamamatsu ORCA AG Black and White CCD camera. Nuclear intensities were measured using a macro designed with ImageJ software.
Fancg-Flag Reconstitution
Mouse
Fancg cDNA sequence was obtained from Ensembl. The cDNA sequence was flanked on both ends by 2 glycine residues and a Flag tag.
Fancg-Flag sequence was ordered as a geneblock from Integrated DNA Technologies (IDT). The sequence was cloned into pMX-IRES-GFP plasmid (Supplementary Figure S1). The orientation and insertion of the geneblock was confirmed using three primer sets (
Table 1). To transduce the
Trp53kd MEFs with
Fancg-Flag cDNA
, HEK293T cells were seeded in a 10 cm dish. The following day, HEK293T cells were transfected with 6 μl X-tremeGENE (Roche), 194 μl of serum free medium (SFM) and incubated for 5 mins at RT. 2 μg of pMX-
Fancg-Flag-IRES-GFP and packaging vector (pCL-Eco) were added in a total of 200 μl Serum Free Medium. The ratio of X-tremeGENE to total DNA was 3:1 while that for the plasmid to pCL-Eco was 3:2. Both X-tremeGENE and plasmid mix were put together and incubated for 30 mins at RT. Following incubation, 400 μl of the final mixture was added dropwise to each well already containing 1.6 ml of complete IMDM medium. These cells were cultured under standard conditions for 48 hrs after which the supernatant containing retroviral particles was collected. Polybrene
® (10 mg/ml) was added to the supernatant at a final concentration of 1 mg/ml. 2 ml of virus supernatant was added to 500,000 MEFs. After 5 days, cells were harvested and sorted for positive GFP expression to be further used for culture.
Statistical Analysis
To assess the statistical significance of our data appropriate tests were performed using GraphPad Prism