The progression of human prostate cancer is a multistep process, beginning with primary tumor formation and ending with metastasis of cancer cells and their growth at secondary tissue sites, such as bone. Although prostate cancer is the most commonly diagnosed visceral neoplasm in men, it typically exhibits slow progression and many more men die with prostate cancer than of it.
1Prostate carcinoma: defining therapeutic objectives and improving overall outcomes.
, 2- Jemal A.
- Siegel R.
- Ward E.
- Hao Y.
- Xu J.
- Thun M.J.
Cancer statistics, 2009.
The development of genetically engineered mouse models of prostate cancer to model these events presents a considerable challenge, because few models are able to mimic this progression series in detail.
3- Pienta K.J.
- Abate-Shen C.
- Agus D.B.
- Attar R.M.
- Chung L.W.
- Greenberg N.M.
- Hahn W.C.
- Isaacs J.T.
- Navone N.M.
- Peehl D.M.
- Simons J.W.
- Solit D.B.
- Soule H.R.
- VanDyke T.A.
- Weber M.J.
- Wu L.
- Vessella R.L.
The current state of preclinical prostate cancer animal models.
One model that has gained considerable attention is prostate-specific deletion of the
Pten tumor suppressor gene, a gene that is commonly disrupted in human prostate cancer.
4- Wang S.
- Gao J.
- Lei Q.
- Rozengurt N.
- Pritchard C.
- Jiao J.
- Thomas G.V.
- Li G.
- Roy-Burman P.
- Nelson P.S.
- Liu X.
- Wu H.
Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer.
Ptenp−/− mice rapidly developed murine prostatic intraepithelial neoplasia (mPIN), which eventually progressed to invasive adenocarcinoma and metastases in 45% of mice by 12 to 29 weeks; however, metastatic disease was not observed by others using a different prostate-specific
Pten mutant allele.
5- Chen Z.
- Trotman L.C.
- Shaffer D.
- Lin H.K.
- Dotan Z.A.
- Niki M.
- Koutcher J.A.
- Scher H.I.
- Ludwig T.
- Gerald W.
- Cordon-Cardo C.
- Pandolfi P.P.
Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis.
The
Ptenp−/− model is now widely used in an attempt to elucidate the mechanistic basis for prostate cancer progression and as a preclinical model to develop therapeutic interventions.
5- Chen Z.
- Trotman L.C.
- Shaffer D.
- Lin H.K.
- Dotan Z.A.
- Niki M.
- Koutcher J.A.
- Scher H.I.
- Ludwig T.
- Gerald W.
- Cordon-Cardo C.
- Pandolfi P.P.
Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis.
, 6- Wang S.
- Garcia A.J.
- Wu M.
- Lawson D.A.
- Witte O.N.
- Wu H.
Pten deletion leads to the expansion of a prostatic stem/progenitor cell subpopulation and tumor initiation.
, 7- Akins E.J.
- Moore M.L.
- Tang S.
- Willingham M.C.
- Tooze J.A.
- Dubey P.
In situ vaccination combined with androgen ablation and regulatory T-cell depletion reduces castration-resistant tumor burden in prostate-specific Pten knockout mice.
, 8- Moussavi M.
- Fazli L.
- Tearle H.
- Guo Y.
- Cox M.
- Bell J.
- Ong C.
- Jia W.
- Rennie P.S.
Oncolysis of prostate cancers induced by vesicular stomatitis virus in PTEN knockout mice.
In the present study, we attempted to improve further on the
Ptenp−/− mouse model of spontaneous prostate cancer using a more sensitive, more specific, and less variable approach to monitoring prostate cancer progression. Toward this end, we introduced the conditional
ROSA26-LSL firefly luciferase allele
11- Safran M.
- Kim W.Y.
- Kung A.L.
- Horner J.W.
- DePinho R.A.
- Kaelin Jr, W.G.
Mouse reporter strain for noninvasive bioluminescent imaging of cells that have undergone Cre-mediated recombination.
and extensively backcrossed mice onto the albino C57BL/6 genetic background (C57BL/6J-
Tyrc-2J/J) and designated these mice C57/Luc/
Ptenp. We found a marked difference in prostate cancer progression in C57/Luc/
Ptenp mice, compared with other
Ptenp−/− mouse models of prostate cancer, indicating a strong influence of the C57BL/6 genetic background. BLI revealed an initial rapid growth phase in the epithelial compartment up to 11 weeks, which was followed by a markedly decreased rate of growth. Progression to microinvasive prostate cancer was rare in 1-year-old animals, and metastatic prostate cancer was never detected. Furthermore, although C57/Luc/
Ptenp−/− mice responded to castration by decreases in bioluminescence signal, they failed to exhibit relapsed growth, a finding in contrast to other report.
10- Liao C.P.
- Zhong C.
- Saribekyan G.
- Bading J.
- Park R.
- Conti P.S.
- Moats R.
- Berns A.
- Shi W.
- Zhou Z.
- Nikitin A.Y.
- Roy-Burman P.
Mouse models of prostate adenocarcinoma with the capacity to monitor spontaneous carcinogenesis by bioluminescence or fluorescence.
Despite the slow disease progression revealed by BLI, another noninvasive imaging modality, magnetic resonance imaging (MRI), indicated progressive increase in prostate volume. Further investigation revealed that this increased signal was derived from fluid retention in the anterior prostate. Furthermore, we documented rapid recruitment of immunosuppressive Gr-1
+CD11b
+ myeloid-derived suppressor cells (MDSCs), highlighting a possible role for inflammation during early stages of disease progression. This model affords the opportunity to identify and evaluate genetic, inflammatory, and environmental modifiers of prostate cancer progression using a sensitive and specific BLI approach to monitor disease progression.
Materials and Methods
Mouse Strains and Genotyping
All animal procedures were performed with approval from the University of Iowa Animal Care and Use Committee and by the Iowa and Purdue institutional review boards. The B6.Cg-Tg(Pbsn-cre)4Prb (Pb-Cre4
+) mice were obtained from the NIH Mouse Models of Human Cancer Consortium, the C;129S4-
Ptentm1Hwu/J (
Ptenfl/fl) mice were obtained from the Jackson Laboratory (Bar Harbor, ME), and the FVB.129S6 (B6)-
Gt(ROSA)26Sortm1(Luc)Kael/J (
ROSA26-LSL-Luc) mice were a kind gift from Dr. William Kaelin of the Dana-Farber Cancer Institute. Mice were intercrossed to combine alleles and then backcrossed to C57BL/6J-
Tyrc-2J/J (Jackson Laboratory) for seven generations. Mice were genotyped for Cre, floxed alleles of
Pten, and luciferase using gene-specific primers, as described previously.
11- Safran M.
- Kim W.Y.
- Kung A.L.
- Horner J.W.
- DePinho R.A.
- Kaelin Jr, W.G.
Mouse reporter strain for noninvasive bioluminescent imaging of cells that have undergone Cre-mediated recombination.
, 12- Wu X.
- Wu J.
- Huang J.
- Powell W.C.
- Zhang J.
- Matusik R.J.
- Sangiorgi F.O.
- Maxson R.E.
- Sucov H.M.
- Roy-Burman P.
Generation of a prostate epithelial cell-specific Cre transgenic mouse model for tissue-specific gene ablation.
, 13- Lesche R.
- Groszer M.
- Gao J.
- Wang Y.
- Messing A.
- Sun H.
- Liu X.
- Wu H.
Cre/loxP-mediated inactivation of the murine Pten tumor suppressor gene.
Strains analyzed in the present study were hemizygous for Pb-Cre and homozygous for
ROSA26-LSL-Luc. Castration of C57/Luc/
Ptenp mice was performed as described previously.
14- Esser A.K.
- Cohen M.B.
- Henry M.D.
Dystroglycan is not required for maintenance of the luminal epithelial basement membrane or cell polarity in the mouse prostate.
Imaging
All BLI was performed with an IVIS100 imaging system (Caliper Life Sciences, Hopkinton, MA) as described previously.
15- Drake J.M.
- Gabriel C.L.
- Henry M.D.
Assessing tumor growth and distribution in a model of prostate cancer metastasis using bioluminescence imaging.
Kinetic imaging studies revealed that the bioluminescence signal showed a broad peak after
D-luciferin (Promega, Madison, WI) injection, with 8 minutes being an optimal imaging time point. Bioluminescence signal kinetics were independent of mouse genotype and age (data not shown). Mice were therefore routinely imaged 8 minutes after luciferin injection, using a 20-cm field of view and an exposure time varying from 1 to 30 seconds. Bioluminescence values were calculated by measuring photon flux (photons/second) in a region of interest surrounding the bioluminescence signal (with the edge of the region of interest defined as 5% maximum signal), using Living Image software version 2.5 (Caliper Life Sciences).
Magnetic resonance imaging was performed on a 4.7-T Varian Unity/INOVA small-bore MRI scanner (Varian, Palo Alto, CA) equipped with a 38-mm-diameter transmit-receive volume radio frequency (RF) coil. T2-weighted images were acquired in all three principal planes (axial, sagittal, and coronal) using a fast spin-echo (FSE) pulse sequence with repetition time TR = 3000 to 4500 ms, echo time TE = 48 ms, echo train length ETL = 8, and averages = 7. A matrix size of 512 × 128 covered a field of view of 72 mm × 32 mm, with 25 to 45 contiguous slices of 1.0 mm thickness. The TR and number of slices depended on the size of the animal and the slice orientation.
RNA, DNA, and Protein Analysis
Tissues from C57/Luc/
Ptenp mice were removed and flash-frozen in liquid nitrogen for subsequent protein and RNA preparation. For RNA extraction, frozen samples were homogenized using a mortar and pestle, suspended into RNeasy lysis buffer (buffer RLT; Qiagen, Valencia, CA), and subjected to RNA extraction under the manufacturer's conditions (RNeasy kit; Qiagen). Purified RNA preparations were subjected to DNase I treatment to remove any contaminating genomic DNA. For protein preparation, tissues were prepared as described previously.
16- Svensson R.U.
- Shey M.R.
- Ballas Z.K.
- Dorkin J.R.
- Goldberg M.
- Akinc A.
- Langer R.
- Anderson D.G.
- Bumcrot D.
- Henry M.D.
Assessing siRNA pharmacodynamics in a luciferase-expressing mouse.
Protein samples (150 μg) were subjected to an
in vitro luciferase activity assay as described previously.
16- Svensson R.U.
- Shey M.R.
- Ballas Z.K.
- Dorkin J.R.
- Goldberg M.
- Akinc A.
- Langer R.
- Anderson D.G.
- Bumcrot D.
- Henry M.D.
Assessing siRNA pharmacodynamics in a luciferase-expressing mouse.
RNA and DNA samples were subjected to quantitative PCR as described previously.
17- Svensson R.U.
- Barnes J.M.
- Rokhlin O.W.
- Cohen M.B.
- Henry M.D.
Chemotherapeutic agents up-regulate the cytomegalovirus promoter: implications for bioluminescence imaging of tumor response to therapy.
For RNA analysis, laser-capture microdissection was performed on 5-μm frozen sections cut using a Leica cryostat microtome (Leica Microsystems, Wetzlar, Germany). Sections were cut from samples embedded in Tissue-Tek optimum cutting temperature compound (Sakura Finetek, Torrance, CA) on charged superfrost glass slides (Fisher Scientific, Waltham, MA), dehydrated using a graded series of ethanol, and air-dried for 30 minutes. For DNA analysis, laser-capture microdissection was performed on paraffin-embedded 7-μm sections cut using a Leica microtome. Prostatic epithelial and stromal cells were collected on Arcturus HS LCM caps (Applied Biosystems, Sunnyvale, CA). RNA was extracted using an Arcturus Picopure RNA isolation kit (Applied Biosystems) and subjected to DNase I treatment to remove any contaminating genomic DNA. DNA was extracted using an Arcturus Picopure DNA isolation kit (Applied Biosystems) according to the manufacturer's instructions.
Histopathology and Immunofluorescence
Prostates were fixed in 4% paraformaldehyde overnight, transferred to 30% ethanol, embedded in paraffin and stained with H&E or Masson's trichrome in accordance with standard procedures. Prostate pathology was assessed according to the criteria established by Shappell et al.
18- Shappell S.B.
- Thomas G.V.
- Roberts R.L.
- Herbert R.
- Ittmann M.M.
- Rubin M.A.
- Humphrey P.A.
- Sundberg J.P.
- Rozengurt N.
- Barrios R.
- Ward J.M.
- Cardiff R.D.
Prostate pathology of genetically engineered mice: definitions and classification The consensus report from the Bar Harbor meeting of the Mouse Models of Human Cancer Consortium Prostate Pathology Committee.
For immunofluorescence, sections were stained with antibodies to phospho S6 (1:200) (S235/S236; Cell Signaling, Danvers, MA), phospho AKT (1:200) (S473, T308; Cell Signaling), and Ki-67 (1:400) (Novocastra, Newcastle, UK), using heated citrate buffer antigen retrieval. For H&E and immunofluorescence analyses, images were taken using an Olympus BX-61 microscope.
Fluorescence-Activated Cell Sorting Analysis of Inflammatory Subsets
Prostate tissue was placed in a solution of 2 μg/mL collagenase D (Roche, Nutley, NJ) in RPMI-1640 medium containing 10% fetal bovine serum. Tissue was minced, placed at 37°C for 1 hour for digestion, and then was passed through a 70-μm filter. Spleens were removed and ground between frosted microscope slides in PBS. Red blood cells were lysed with ACK buffer and passed through a 70-μm filter. Single cell suspensions were then incubated with TruStain FcX antibody (BioLegend, San Diego, CA) and stained with directly conjugated antibodies (eBioscience, San Diego, CA; BioLegend) according to the manufacturer's instructions. Flow-cytometric analysis was performed on a FACSCanto system (BD Biosciences, Franklin Lakes, NJ) and data were analyzed using FlowJo software version 7.6.2 (Tree Star, Ashland, OR).
MDSC Activation Assays
For analysis of arginase-1 (Arg1) and inducible nitric oxide synthase (iNOS) expression analysis, total RNA was prepared from fluorescence-activated cell-sorted CD45+Gr-1+CD11b+ cells using an RNeasy kit (Qiagen). cDNA was synthesized using a qScript Flex cDNA synthesis kit (Quanta BioSciences, Gaithersburg, MD). Quantitative RT-PCR was performed using TaqMan primer and probe sets for mouse Arg1, iNOS, and 18S rRNA (Applied Biosystems, Carlsbad, CA). Relative mRNA expression was calculated as 2 − (Ct of Arg1 or iNOS − Ct of 18S rRNA), where Ct is the threshold cycle value. Data were normalized to 18S RNA and are representative of three independent experiments.
For in vitro suppression assays, CD45+Gr-1+CD11b+ cells were pooled from four mice per group. Cells were sorted from tissues using an iCyt Reflection cell sorter (iCyt, Champaign, IL) and were seeded at 1 × 105 per well. Thy1.1+ OT-I cells were preactivated for 24 to 48 hours with SIINFEKL (1 mg/mL), purified by Fico/Lite gradient medium (Atlanta Biologicals, Lawrenceville, GA), and added at 1 × 105 cells per well. Bromodeoxyuridine (BrdU) was added directly to OT-I cells at the time of incubation with MDSC and cells were harvested for analysis of proliferation after 12 hours. To evaluate suppression, the percentage of BrdU+Thy1.1+ cells was analyzed by flow cytometry. The percentage suppression of proliferation was calculated as [1 − (proliferation with MDSC/proliferation without MDSC)] × 100.
Discussion
In the present study, we have developed a new model of prostate cancer, C57/Luc/
Ptenp mice. Prostate-specific deletion of
Pten was first reported in 2003,
4- Wang S.
- Gao J.
- Lei Q.
- Rozengurt N.
- Pritchard C.
- Jiao J.
- Thomas G.V.
- Li G.
- Roy-Burman P.
- Nelson P.S.
- Liu X.
- Wu H.
Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer.
and since then has become a widely used mouse model of prostate cancer. We developed C57/Luc/
Ptenp mice in an effort to improve on this model. We found that bioluminescence signal intensity in C57/Luc/
Ptenp mice was significantly greater than in other comparable models,
10- Liao C.P.
- Zhong C.
- Saribekyan G.
- Bading J.
- Park R.
- Conti P.S.
- Moats R.
- Berns A.
- Shi W.
- Zhou Z.
- Nikitin A.Y.
- Roy-Burman P.
Mouse models of prostate adenocarcinoma with the capacity to monitor spontaneous carcinogenesis by bioluminescence or fluorescence.
, 19- Hsieh C.L.
- Xie Z.
- Yu J.
- Martin W.D.
- Datta M.W.
- Wu G.J.
- Chung L.W.
Non-invasive bioluminescent detection of prostate cancer growth and metastasis in a bigenic transgenic mouse model.
, 20- Seethammagari M.R.
- Xie X.
- Greenberg N.M.
- Spencer D.M.
EZC-prostate models offer high sensitivity and specificity for noninvasive imaging of prostate cancer progression and androgen receptor action.
was almost exclusively restricted to prostate epithelial cells, and was correlated with the proliferative capacity of the prostatic epithelium. Although BLI of 3- to 11-week-old C57/Luc/
Ptenp−/− mice revealed rapid growth of the prostatic epithelium, subsequent epithelial expansion was slow. MRI revealed progressive increases in prostate volume, which was attributed to excessive fluid retention and stromal responses in AP rather than to solid tumor growth, warranting caution when using MRI to analyze prostate growth in tumor-prone mice. Furthermore, we found a marked difference in prostate cancer progression in C57/Luc/
Ptenp−/− mice, compared with previous studies in
Ptenp−/− mice. Invasive prostate cancer was rare even in 1-year-old animals, and we did not detect metastatic disease.
Several studies have described the detection of metastatic prostate cancer in
Ptenp−/− animals,
4- Wang S.
- Gao J.
- Lei Q.
- Rozengurt N.
- Pritchard C.
- Jiao J.
- Thomas G.V.
- Li G.
- Roy-Burman P.
- Nelson P.S.
- Liu X.
- Wu H.
Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer.
, 10- Liao C.P.
- Zhong C.
- Saribekyan G.
- Bading J.
- Park R.
- Conti P.S.
- Moats R.
- Berns A.
- Shi W.
- Zhou Z.
- Nikitin A.Y.
- Roy-Burman P.
Mouse models of prostate adenocarcinoma with the capacity to monitor spontaneous carcinogenesis by bioluminescence or fluorescence.
, 30- Ma X.
- Ziel-van der Made A.C.
- Autar B.
- van der Korput H.A.
- Vermeij M.
- van Duijn P.
- Cleutjens K.B.
- de Krijger R.
- Krimpenfort P.
- Berns A.
- van der Kwast T.H.
- Trapman J.
Targeted biallelic inactivation of Pten in the mouse prostate leads to prostate cancer accompanied by increased epithelial cell proliferation but not by reduced apoptosis.
, 31- Burton J.B.
- Johnson M.
- Sato M.
- Koh S.B.
- Mulholland D.J.
- Stout D.
- Chatziioannou A.F.
- Phelps M.E.
- Wu H.
- Wu L.
Adenovirus-mediated gene expression imaging to directly detect sentinel lymph node metastasis of prostate cancer.
and all have reported highly penetrant invasive adenocarcinomas, albeit with variable latency.
5- Chen Z.
- Trotman L.C.
- Shaffer D.
- Lin H.K.
- Dotan Z.A.
- Niki M.
- Koutcher J.A.
- Scher H.I.
- Ludwig T.
- Gerald W.
- Cordon-Cardo C.
- Pandolfi P.P.
Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis.
, 23- Zhang W.
- Zhu J.
- Efferson C.L.
- Ware C.
- Tammam J.
- Angagaw M.
- Laskey J.
- Bettano K.A.
- Kasibhatla S.
- Reilly J.F.
- Sur C.
- Majumder P.K.
Inhibition of tumor growth progression by antiandrogens and mTOR inhibitor in a Pten-deficient mouse model of prostate cancer.
, 24- Lin H.K.
- Chen Z.
- Wang G.
- Nardella C.
- Lee S.W.
- Chan C.H.
- Yang W.L.
- Wang J.
- Egia A.
- Nakayama K.I.
- Cordon-Cardo C.
- Teruya-Feldstein J.
- Pandolfi P.P.
Skp2 targeting suppresses tumorigenesis by Arf-p53-independent cellular senescence.
, 25- Trotman L.C.
- Niki M.
- Dotan Z.A.
- Koutcher J.A.
- Di Cristofano A.
- Xiao A.
- Khoo A.S.
- Roy-Burman P.
- Greenberg N.M.
- Van Dyke T.
- Cordon-Cardo C.
- Pandolfi P.P.
Pten dose dictates cancer progression in the prostate.
, 26- Nardella C.
- Carracedo A.
- Alimonti A.
- Hobbs R.M.
- Clohessy J.G.
- Chen Z.
- Egia A.
- Fornari A.
- Fiorentino M.
- Loda M.
- Kozma S.C.
- Thomas G.
- Cordon-Cardo C.
- Pandolfi P.P.
Differential requirement of mTOR in postmitotic tissues and tumorigenesis.
, 30- Ma X.
- Ziel-van der Made A.C.
- Autar B.
- van der Korput H.A.
- Vermeij M.
- van Duijn P.
- Cleutjens K.B.
- de Krijger R.
- Krimpenfort P.
- Berns A.
- van der Kwast T.H.
- Trapman J.
Targeted biallelic inactivation of Pten in the mouse prostate leads to prostate cancer accompanied by increased epithelial cell proliferation but not by reduced apoptosis.
, 32- Backman S.A.
- Ghazarian D.
- So K.
- Sanchez O.
- Wagner K.U.
- Hennighausen L.
- Suzuki A.
- Tsao M.S.
- Chapman W.B.
- Stambolic V.
- Mak T.W.
Early onset of neoplasia in the prostate and skin of mice with tissue-specific deletion of Pten.
To our knowledge, the present study is the first to report the tumor phenotype in cohorts of prostate-specific
Ptenp−/− animals extensively backcrossed to the C57BL/6 genetic background. This model does not recapitulate the pace, extent, or variability of disease progression associated with prostate-specific
Pten deletion on mixed 129 genetic backgrounds (ie, C57BL/6;DBA;129/BALB/c;FVB/N,
10- Liao C.P.
- Zhong C.
- Saribekyan G.
- Bading J.
- Park R.
- Conti P.S.
- Moats R.
- Berns A.
- Shi W.
- Zhou Z.
- Nikitin A.Y.
- Roy-Burman P.
Mouse models of prostate adenocarcinoma with the capacity to monitor spontaneous carcinogenesis by bioluminescence or fluorescence.
129/C57BL/6,
32- Backman S.A.
- Ghazarian D.
- So K.
- Sanchez O.
- Wagner K.U.
- Hennighausen L.
- Suzuki A.
- Tsao M.S.
- Chapman W.B.
- Stambolic V.
- Mak T.W.
Early onset of neoplasia in the prostate and skin of mice with tissue-specific deletion of Pten.
129;C57BL/6;DBA;BALB/c,
4- Wang S.
- Gao J.
- Lei Q.
- Rozengurt N.
- Pritchard C.
- Jiao J.
- Thomas G.V.
- Li G.
- Roy-Burman P.
- Nelson P.S.
- Liu X.
- Wu H.
Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer.
, 25- Trotman L.C.
- Niki M.
- Dotan Z.A.
- Koutcher J.A.
- Di Cristofano A.
- Xiao A.
- Khoo A.S.
- Roy-Burman P.
- Greenberg N.M.
- Van Dyke T.
- Cordon-Cardo C.
- Pandolfi P.P.
Pten dose dictates cancer progression in the prostate.
and 129;FVB
30- Ma X.
- Ziel-van der Made A.C.
- Autar B.
- van der Korput H.A.
- Vermeij M.
- van Duijn P.
- Cleutjens K.B.
- de Krijger R.
- Krimpenfort P.
- Berns A.
- van der Kwast T.H.
- Trapman J.
Targeted biallelic inactivation of Pten in the mouse prostate leads to prostate cancer accompanied by increased epithelial cell proliferation but not by reduced apoptosis.
). Prior studies have clearly demonstrated that
Pten-dependent tumorigenesis is influenced by genetic background,
33- Freeman D.
- Lesche R.
- Kertesz N.
- Wang S.
- Li G.
- Gao J.
- Groszer M.
- Martinez-Diaz H.
- Rozengurt N.
- Thomas G.
- Liu X.
- Wu H.
Genetic background controls tumor development in PTEN-deficient mice.
which is also known to affect prostate gene expression in mice.
34- Bianchi-Frias D.
- Pritchard C.
- Mecham B.H.
- Coleman I.M.
- Nelson P.S.
Genetic background influences murine prostate gene expression: implications for cancer phenotypes.
Blando et al
35- Blando J.
- Portis M.
- Benavides F.
- Alexander A.
- Mills G.
- Dave B.
- Conti C.J.
- Kim J.
- Walker C.L.
PTEN deficiency is fully penetrant for prostate adenocarcinoma in C57BL/6 mice via mTOR-dependent growth.
recently reported that
Pten deficiency is fully penetrant for prostate adenocarcinoma in the anterior prostate of constitutive
Pten+/− animals on an extensively backcrossed C57BL/6 genetic background. This finding is most surprising in light of our results, in which there was little progression beyond focal mPIN in the C57/Luc/
Ptenp+/− mice. There may be a different cell of origin in the constitutive
Pten knockout mouse prostate, compared with tissue-specific knockout mice using Pb-Cre4. Other studies are in accord with our findings of mild disease progression in the C57BL/6 background. TRAMP mice on a C57BL/6 genetic background frequently survive to 36 to 40 weeks of age, and sometimes up to 52 weeks of age, whereas those on a C57BL/6;FVB F1 background never survive past 33 weeks of age.
36- Gingrich J.R.
- Barrios R.J.
- Foster B.A.
- Greenberg N.M.
Pathologic progression of autochthonous prostate cancer in the TRAMP model.
However, genetic background is likely to collaborate with the specific mutations used to drive prostate cancer in mouse models.
37- Xu Q.
- Majumder P.K.
- Ross K.
- Shim Y.
- Golub T.R.
- Loda M.
- Sellers W.R.
Identification of prostate cancer modifier pathways using parental strain expression mapping.
, 38- Kim M.J.
- Bhatia-Gaur R.
- Banach-Petrosky W.A.
- Desai N.
- Wang Y.
- Hayward S.W.
- Cunha G.R.
- Cardiff R.D.
- Shen M.M.
- Abate-Shen C.
Nkx3.1 mutant mice recapitulate early stages of prostate carcinogenesis.
Because in this model disease progression can be assessed noninvasively, the C57/Luc/
Ptenp mice described here could be a valuable tool for forward genetic analysis of modifier loci.
The present study also revealed that expansion of the epithelial compartment in C57/Luc/
Ptenp−/− mice animals is biphasic, with an initial rapid phase followed by a slow phase. During the rapid phase, we found that the immediate increase in bioluminescence signal intensity from 3 to 6 weeks is due to the development of mPIN in DP and LP, compared with 6 to 11 weeks, when increases in bioluminescence are correlated with mPIN development in VP and AP. During the slow phase from 11 weeks onward, we did not see pronounced development of further mPIN. Moreover, proliferative levels of the epithelium were substantially reduced, compared with the rapid phase. Prior studies indicate that this apparent growth arrest may be due to p53- or p27-induced senescence.
5- Chen Z.
- Trotman L.C.
- Shaffer D.
- Lin H.K.
- Dotan Z.A.
- Niki M.
- Koutcher J.A.
- Scher H.I.
- Ludwig T.
- Gerald W.
- Cordon-Cardo C.
- Pandolfi P.P.
Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis.
, 39- Majumder P.K.
- Grisanzio C.
- O'Connell F.
- Barry M.
- Brito J.M.
- Xu Q.
- Guney I.
- Berger R.
- Herman P.
- Bikoff R.
- Fedele G.
- Baek W.K.
- Wang S.
- Ellwood-Yen K.
- Wu H.
- Sawyers C.L.
- Signoretti S.
- Hahn W.C.
- Loda M.
- Sellers W.R.
A prostatic intraepithelial neoplasia-dependent p27 Kip1 checkpoint induces senescence and inhibits cell proliferation and cancer progression.
However, it is striking that the biphasic kinetics of bioluminescence signal are independent of
Pten status, suggesting that the majority of epithelial tumor growth occurs during the development and maturation phase of the prostate. This raises the possibility that developmental mechanisms in the prostate may trigger a senescence program driven by
Pten disruption. Indeed, the developmental timing of
Pten deletion has been shown to affect the disease phenotype.
40- Luchman H.A.
- Benediktsson H.
- Villemaire M.L.
- Peterson A.C.
- Jirik F.R.
The pace of prostatic intraepithelial neoplasia development is determined by the timing of Pten tumor suppressor gene excision.
, 41- Ratnacaram C.K.
- Teletin M.
- Jiang M.
- Meng X.
- Chambon P.
- Metzger D.
Temporally controlled ablation of PTEN in adult mouse prostate epithelium generates a model of invasive prostatic adenocarcinoma.
We also found that prostate tumors from C57/Luc/
Ptenp−/− mice failed to proliferate in the castrate state for up to 1 year, consistent with the overall diminished disease phenotype in this model. However, bioluminescence signal maintained at a constant level, approximately one order of magnitude above baseline only 4 weeks after castration, suggesting that a population of castrate-resistant cells that were already present in C57/Luc/
Ptenp−/− prostate tumors were selected for during androgen deprivation. Accordingly, Gao et al
42- Gao H.
- Ouyang X.
- Banach-Petrosky W.A.
- Shen M.M.
- Abate-Shen C.
Emergence of androgen independence at early stages of prostate cancer progression in Nkx3.1; pten mice.
demonstrated that prostate epithelial cells from
Nkx3.1;
Pten mutant mice were capable of surviving in the absence of androgens well before they displayed an overt cancer phenotype, suggesting the early emergence of a castrate-resistant phenotype.
The tumor-associated stroma is a complex microenvironment that consists of a number of extracellular matrix proteins, growth factors, fibroblasts, and immune cells. Stromal expansion in C57/Luc/
Ptenp−/− animals was accompanied by increased collagen deposition, as reported previously,
4- Wang S.
- Gao J.
- Lei Q.
- Rozengurt N.
- Pritchard C.
- Jiao J.
- Thomas G.V.
- Li G.
- Roy-Burman P.
- Nelson P.S.
- Liu X.
- Wu H.
Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer.
but also by enhanced recruitment of Gr-1
+CD11b
+ MDSCs. MDSCs have been shown to accumulate at tumor sites in experimental animals of cancer,
43- Youn J.I.
- Nagaraj S.
- Collazo M.
- Gabrilovich D.I.
Subsets of myeloid-derived suppressor cells in tumor-bearing mice.
, 44- Zhao F.
- Obermann S.
- von Wasielewski R.
- Haile L.
- Manns M.P.
- Korangy F.
- Greten T.F.
Increase in frequency of myeloid-derived suppressor cells in mice with spontaneous pancreatic carcinoma.
but their accumulation in mouse models of spontaneous prostate cancer has not been assessed to date. We have shown that prostates of C57/Luc/
Ptenp−/− mice display a progressive increase in recruitment of Gr-1
+CD11b
+ cells (which are identified as MDSCs, based on functional studies at 4 months). Furthermore, Gr-1
+CD11b
+ cells isolated from the prostates expressed high levels of Arg1 and iNOS and were able to more effectively suppress T-cell proliferation
in vitro than Gr-1
+CD11b
+ cells from spleens of the same animals. A recently published model of abacterial prostatitis showed that the immunosuppressive function of MDSC is limited to the inflammatory site.
45- Haverkamp J.M.
- Crist S.A.
- Elzey B.D.
- Cimen C.
- Ratliff T.L.
In vivo suppressive function of myeloid-derived suppressor cells is limited to the inflammatory site.
We did not, in the present study, determine the relative contribution of MDSCs in prostate cancer progression in these animals, but previous studies have highlighted a crucial role for MDSCs in early cancer development.
46- Tu S.
- Bhagat G.
- Cui G.
- Takaishi S.
- Kurt-Jones E.A.
- Rickman B.
- Betz K.S.
- Penz-Oesterreicher M.
- Bjorkdahl O.
- Fox J.G.
- Wang T.C.
Overexpression of interleukin-1beta induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice.
Although speculative, the present data demonstrating diminished prostate cancer progression in C57/Luc/
Ptenp−/− mice suggest that MDSC function may be linked to initial tumor development but not to progression to an invasive phenotype. Although the C57/Luc/
Ptenp−/− mouse reported here has a much milder phenotype than other prostate-specific
Pten mutant models, this model may nonetheless provide a very useful background for evaluating genetic and environmental factors that convert an indolent to an aggressive disease—a process that remains a central question for prostate cancer in humans.
Article info
Publication history
Published online: May 09, 2011
Accepted:
March 31,
2011
Footnotes
Supported by pilot grants from the University of Iowa Cancer and Aging Program (5 P20 CA103672), by the Center for Health Effects of Environmental Contamination, and by NIH grants R21-CA137490 and RC1-ES018097 (M.D.H.) and R21-CA154126 (T.L.R. and M.D.H.).
Supplemental material for this article can be found at http://ajp.amjpathol.org or at doi: 10.1016/j.ajpath.2011.03.014.
Copyright
© 2011 American Society for Investigative Pathology. Published by Elsevier Inc.