Abstract
Prostate cancer (PCa) is a significant health concern for men worldwide and is particularly prevalent in the United States. It is a complex disease presenting different molecular subtypes and varying degrees of aggressiveness. Transgenic/genetically engineered mouse models (GEMMs) greatly enhanced our understanding of the intricate molecular processes that underlie PCa progression and advancement and have offered valuable insights into potential therapeutic targets for this disease. The integration of whole-exome and whole-genome sequencing, along with expression profiling, has played a pivotal role in advancing GEMMs by facilitating the identification of genetic alterations driving PCa development. This review focuses on genetically modified mice classified into the first and second generations of PCa models. We summarize whether models created by manipulating the function of specific genes replicate the consequences of genomic alterations observed in human PCa, including early and later disease stages. We discuss cases where GEMMs did not fully exhibit the expected human PCa phenotypes and possible causes of the failure. Here, we summarize the comprehensive understanding, recent advances, and the strengths and limitations of the GEMMs in advancing our insights into PCa, offering genetic and molecular perspectives for developing novel GEMM models.
Keywords: Prostate cancer, Transgenic mice, GEMMs, Genes, Genomic alterations
1. Introduction
Prostate cancer (PCa) is the second most frequently diagnosed non-cutaneous cancer and the fifth leading cause of cancer-related death in men globally. In the United States, it is a prevalent form of cancer in men, accounting for approximately 14% of all new cancer cases [1][2]. Radical prostatectomy remains a gold standard for treating localized PCa, and androgen deprivation therapy (ADT) is the backbone of systemic therapy for those who experience disease relapse after local therapy or for those with metastatic disease [3][4][5]. Poor prognoses persist in patients with advanced disease, attributed to factors like age, race, oxidative stress, inflammation, disease heterogeneity, drug resistance, and distant metastases [6][7]. As a result, the five-year disease-specific survival rate for metastatic disease stands at approximately 32.3% [1].
Creating genetically engineered mouse models (GEMMs) is crucial for studying diseases like cancers, allowing researchers to mimic human pathology [8][9]. During the 1980s, independent researchers unknowingly developed the first transgenic mice expressing dominant oncogenes (oncomice), highlighting concurrent progress in genetic engineering [10]. A breakthrough in this area was the creation of tissue-specific promoters and “Cre-lox” technology [11][12]. In cancer biology, there are three major types of genetic modification: "knock-in", involving the insertion of a transgene or modified allele; the “knock-out”, which entails the targeted inactivation of a specific gene or DNA segment to assess its function; and the “conditional and/or inducible knock-out”, which allows for spatially or temporally specific knockout [13]. Several strategies are utilized to integrate a transgene into the mouse genome to generate transgenic and gene knockout/knockin mouse models [14][15]. Major approach to introduce germline genetic modifications in a mouse include zygote route using pronuclear microinjection, and embryonic stem (ES) cell mediated gene -targeted transgene approach [16]. In the standard transgenic approach, the target transgene is injected directly into the pronucleus of murine embryos at the pronuclear stage [17][18]. The modified embryos are then transferred to surrogate mothers for gestation and development. Integration of the transgene into the genome typically occurs randomly. In recent years, exciting advancements in technology have been applied to the zygote route include the use of Sleeping Beauty transposons, ensuring reliable germline transgenesis[19]. On the other hand, in the gene-targeted transgenic approach, the transgene is introduced into ES cells [13][16]. The modified ES cells are then injected into blastocysts, which are subsequently implanted into surrogate mothers. This method allows for more precise targeting of the transgene to specific genomic loci. In addition to these, several other methods have been utilized in genetic research such as somatic cell nuclear transfer, retroviral -mediated transgenesis, homologous recombination in ES cells, sperm- and testicular-mediated gene transfer, Cre-lox mediated gene targeting, RNAi-mediated gene targeting, and Gene editing tool [15]. The advent of gene editing tools represents the most significant revolution in genetic research. This includes technologies like zinc-finger nucleases, transcription activator–like effector nucleases (TALENs), and clustered regularly interspersed short palindromic repeats (CRISPRs)/CRISPR-associated (Cas) systems [20][21][22]. The CRISPR/Cas9 system allows for precise insertion of transgenes into specific genomic locations in mice, ensuring high efficiency and control over integration sites, facilitating the generation of knock-in transgenic mice. Each of these strategies has its advantages and limitations, and the choice of method depends on factors such as the desired level of precision, efficiency, and experimental requirements.
Mouse modeling PCa is complex due to fundamental disparities in prostate biology and tumorigenesis among mice and humans. In comparison, one in six human males is at risk of developing PCa during their lifetime, whereas spontaneous PCa development is rare in mice, the translatability of findings [23]. Another challenge is that the mouse prostate is not a single organ but consists of four distinct lobes. In view of this, there are concerns regarding which mouse prostate lobe(s) resemble the human prostate the most. Further complicating the interpretation of experimental results is the variation in the stroma surrounding mouse and human prostate lobes [24][25]. This review aims to provide a comprehensive overview of the historical development and phenotypic description of the most widely utilized transgenic/genetically engineered mouse models of PCa that replicate the genomic variations that appear in human PCa. Moreover, the review also highlights the challenges/limitations of using mice models that are first- or second-generation derived. We have extensively discussed each model's conceptual grasp, current developments, and clinically pertinent issues and highlighted the strengths and challenges/limitations of first- or second-generation derived mice models to understand pathobiology and molecular aspects of PCa.
2. Comparative anatomy of mouse vs. Human prostate and morphological progression model of PCa
There are notable differences between the overall size, structure, and complexity of the glandular architecture of the human and mouse prostate [26]. The prostate gland in humans consists of three distinct histological zones: the peripheral (PZ), transitional (TZ), and central (CZ) zones. The PZ is the largest and most common site of PCa; the TZ surrounds the urethra and is a common site for developing benign prostatic hyperplasia (BPH), and the CZ surrounds the ejaculatory ducts (Figure 1A). On the other hand, the prostate gland in mice is divided into four lobes: the dorsal (DP), lateral (LP), anterior (AP), and ventral (VP) lobes, with the dorsal and lateral combined to form the dorsolateral lobe (DLP) (Figure 1A) [26][27]. Both human and mouse prostate epithelium contain luminal, basal, intermediate, and neuroendocrine cell populations (Figure 1 B, C); there is a notable difference in the arrangement of basal cells. The basal cell layer in human prostates appears continuously in histological sections, whereas in mouse prostates, basal cells connect through long cytoplasmic processes. Resultant a 1:1 ratio of basal to luminal cells in humans, while this ratio is ~1:7 in mice. The stromal tissue surrounding the epithelium varies between the two species [28].
Figure 1. Anatomy/ morphological structure of the murine and human prostate gland along with critical genetic/molecular alterations and schematic illustration of PCa evolution:
(A) The anatomical representation of the mouse and the human body and the location of the prostate gland. In mice, the prostate gland is located just below the bladder and surrounds the urethra. In humans, it is located in the pelvis, between the bladder and the rectum, and surrounds the urethra. (B) Diagram depicting the architecture of mouse and human prostate gland and adjacent structure. In mice, the prostate gland consists of four major lobes - the anterior, dorsal, lateral, and ventral lobes (left). In humans, the prostate gland is divided into three major prostatic zones - the peripheral zone, central zone, and transitional zone(right) (C) Representation of different cell types in the prostate. Histologically, it consists of secretory luminal, basal, intermediate, and neuroendocrine cells. The architecture of the prostate gland in both mice and humans consists of glandular epithelium embedded in the fibromuscular stroma. The prostatic epithelium is separated from the stroma by a basal lamina. It also comprises ducts and acini, which contain several cell types: smooth muscle cells, fibroblast, endothelial, and neurons. (D) A series of specific genetic and molecular alterations are involved in different stages of the PCa progression model, including PIN, localized adenocarcinoma, and invasive carcinoma. These alterations have been represented by deletion or inactivating mutation of the tumor-suppressor genes, overexpression of protooncogene, modulation of chemokines and pathway regulatory proteins, telomerase activation, and other molecular events as per disease stage (PIN, localized adenocarcinoma and metastasis). (E) Morphologic features of normal prostatic epithelium consisting of basal, luminal, and neuroendocrine cells. PCa progression occurs from either the luminal or basal cells of normal epithelium. At first, the development of premalignant lesions known as PIN has the potential to transform into low or high-grade carcinoma. Eventually, it becomes a metastatic disease spreading to the lymph nodes, bone, liver, and lungs via the circulation system. (F) The percentage bar graph presentation of human genomic alterations (Multiple alteration, fusion, truncating mutation, deletion, missense mutation, and amplification) concerning human metastasis and CRPC [9].
Notably, human and mouse tumors experience common biological processes driven by orthologous genetic events in their malignant evolution and exhibit noteworthy similarities in anatomy and morphology characteristics [29][30]. Regarding cellular populations, it has been observed that mouse distal and proximal luminal cells exhibit the closest similarity to human acinar and ductal populations, respectively. Nonetheless, here are some key points of comparison of the mouse and human prostate: (i) Lobes: Initially, VP in mice is often considered to be the most similar to the human peripheral zone, which is the region where most prostate cancers originate. Using single-cell analysis by Crowley et al., the similarity of cell composition between the mouse lateral prostate and the human peripheral zone was recently confirmed. The study showed that mouse distal and proximal luminal cells are most similar to human acinar and ductal populations. A periurethral population (PrU) that shares basal and luminal features is conserved between species [31]. (ii) Histology: Both mouse and human prostates comprise glandular tissue. They consist of epithelial cells arranged in glandular structures and are surrounded by stromal cells [32]. To account for differences and similarities in human and mouse prostate characteristics, investigators should use the ‘Bar Harbor Classification System’ when characterizing new GEMMs or undertaking experimental interventions that may impact the phenotype or natural history of lesion progression in existing models [21].
PCa is a multifaceted disease that develops, progresses, and advances through various genetic mutations, signaling pathway modulation, and epigenetic modifications that result in the dysregulation of vital cellular functions (Figure 1D). The progression model of PCa is represented (Figure 1E). Recent reports have summarized the human mutational and genomic alterations involved that lead to more advanced phenotypes often associated with castration-resistance prostate cancer (CRPC) and metastasis (Figure 1F) [33][9].
3. First-generation transgenic mouse models of PCa
3.1. Diverse promoters
A set of regulatory elements derived from different genes and regions have been effectively utilized to direct heterologous genes in prostatic epithelial cells of transgenic mice. These regulatory elements include (i) promoter/enhancer region from the rat C3(1) gene, (ii) androgen-sensitive rat probasin (PB), (iii) human prostate-specific antigen (PSA) gene and (iv) mouse mammary tumor virus (MMTV) long terminal repeat (LTR) [34][35][36][37]. The most commonly used promoter for developing transgenic mice lines is from the rat PB (PB), which is sufficient to drive androgen-responsive target expression in the prostatic epithelium [38][39][40]. PB promoter includes a minimal (−426 bp to +28 bp), PB DNA segment (also named PB), a larger (−11.5 kb to +28 bp) PB DNA segment (also named LPB), and a composite minimal PB DNA segment (also called ARR2PB) [35][39][41]. The ARR2PB-modified promoter was engineered by combining the minimal rat PB with an element of its enhancer region encompassing the two androgen-responsive elements (AREs) [35].
3.2. Viral oncogene-driven transgenic mouse models
First-generation transgenic mouse models have widely influenced PCa research and have provided a foundation for developing more advanced models (Figure 2A, B). The T antigen is an oncogenic viral gene in the genetic material of viruses like Simian virus 40 (SV40) and Polyoma virus [42] [43]. In the SV40 early region, two primary oncogenic proteins are prominent: the large tumor T antigen (Tag) and the small t antigen [44]. The large T antigen is responsible for cellular transformation via inactivating two essential tumor suppressor proteins, including p53 and RB [45][46][47]. The p53 transcription factor regulates the cell cycle and promotes apoptosis in cells under stress or damage. At the same time, RB controls the cell cycle by preventing cells from entering the S phase until they undergo proper growth control (Figure 2C). Similarly, the small t antigen interacts with the serine/threonine phosphatase PP2A and stimulates mitogen-activated protein kinase, which regulates several intracellular proteins implicated in various aspects of cellular transformation, including cell growth, proliferation, and survival thereby contributing to cancer development [48]. Therefore, the SV40 early region is considered an oncogenic "sledgehammer"[24].
Figure 2. Strategy for establishing a transgenic mouse model, traditional target vectors, and mode of the molecular action of SV-40 Tag:
(A) Standered transgenic approach [17][18]: Donor female mice were superovulated and allowed to mate overnight with stud male mice (1:1). Embryos were retrieved from the oviducts of donor females. Simultaneously, the preparation of the transgene construct and production of pseudopregnant recipient females were carried out. A transgene DNA fragment was injected into the male pronucleus of murine pronuclear stage embryos from the donor mice. Viable injected embryos were subsequently transferred to pseudopregnant recipient mice. Offspring were screened for the presence of the transgene through genotyping, and founders were identified. (B) Several promoters, including PB and non-PB systems, were used to generate transgenic mice. (C) Molecular mechanism of SV40-early region (large-T antigen) via targeting of tumor suppressor genes RB and P53. Specifically, the pRB-related protein family (pRB, p107, and p130/pRb2) generally binds to transcription factor E2F in early G1 of the cell cycle to prevent the expression of growth-stimulatory genes. The presence of SV40 T-ag disrupts this interaction, leading to the release of E2F and activation of these genes. In addition, in general, the role of P53 is to sense DNA damage and halt the cell cycle to allow for DNA repair or initiate apoptosis. The SV40 T-ag binds to P53 and blocks its function, allowing cells with genetic damage to survive and replicate.
The Male C3(1)-Tag mice are the first transgenic mouse line reported for studying PCa [34][49]. Various transgenic mouse lines have been created using different promoters, such as cryptdin 2/SV40 large-T/small-t, fetal gamma-globin/SV40 large-T/small-t (Gγ-globin-Tag), gp-91phox/SV40 large-T/small-t, MMTV/kgf, and others [50][51][52][53][54]. These lines exhibit distinct profiles of disease progression, as described in Table 1. A challenge with these transgenic mice is the risk of unintended off-target effects, resulting in transgene expression in other tissues due to the random integration of transgenes into the host genome.
Table 1.
Transgenic prostate cancer mouse model using non-probasin and prostate-specific probasin promoters.
Mouse Model | Features | References |
---|---|---|
C3 (1)-Tag | Develops prostatic epithelial hyperplasia in
12 weeks. Progression of prostate carcinoma with neuroendocrine differentiation within 28-44 weeks. Other infected tissues include the mammary gland, testis, and salivary gland. Metastatic sites infrequently to the lung. |
[34] [155][156] |
C3(1)/Py-MT | Develops prostatic lesions in both the ventral and dorsolateral lobe ranging from dysplasia to invasive carcinoma. | [157] |
C3(1)/bcl-2 | Proliferative lesions in the prostate
epithelium in the ventral lobe, but unable to develop carcinoma and
metastasis. Lesions were characterized by enhanced epithelial and stromal cell populations, which is similar to human BPH (benign prostatic hyperplasia). Transgene expression was also seen in the testis and uterus without phenotypic anomalies. |
[158] |
TRAMP (PB (−426 to +28bp) / SV40 early region) | Display mild to extensive hyperplasia with
cribriform structures by 8-12 weeks, PIN within 18-24 weeks, and
progression to carcinoma arises by 28 weeks. Develops poorly differentiated carcinomas with neuroendocrine phenotypes by 24 weeks. Metastasis frequently occurs in regional lymph nodes and lungs and less often in bone, kidney, and adrenal gland by 24-30 weeks. Castration-resistant phenotype. |
[40][56] [61] [55][59][60] |
LADY (PB (−11.5 to +28 kb) /SV40 Tag) | Develops prostatic epithelial hyperplasia by
10 weeks. Progressive lesions of high-grade dysplasia and poorly undifferentiated adenocarcinoma by 20 weeks. Commonly metastasize to regional lymph nodes, liver, and lung. Androgen-dependent tumor growth. Neuroendocrine phenotype. |
[41][70][71] |
Cryptdin 2/ SV 40 large-T/small-t | Develops rapidly progressive PCa with
neuroendocrine features, followed by PIN to metastasis by 12 weeks, even
without androgen stimulation. Commonly metastasize to lymph nodes, liver, lung, and bone by 16 weeks. |
[50] |
Fetal gamma globin/ SV 40 large-T/small-t (Gγ-globin-Tag) | Develops invasive carcinoma with
neuroendocrine features by 16-20
weeks. Androgen-independent tumors are considered an accurate model for CRPC. Metastasize to the adrenal gland, lung, lymph nodes, bone, thymus, and intracapsular tissue of the neck and shoulders by 12-16 weeks. |
[51][52] |
gp-91phox/SV40 large-T/small-t | Develops invasive carcinoma with
neuroendocrine features. Metastasize to surrounding tissue. Used for studying neuroectodermal malignancies. |
[53] |
MMTV/kgf | Develops prostatic epithelial hyperplasia in
the ventral and dorsolateral lobe, seminal vesicles, and vas
deferens. No invasive carcinoma, no metastasis. Transgene expression is noticed in the salivary and mammary glands of males. |
[54] |
MMTV/int2/fgf3 | Develop enlarged ampullary glands, seminal vesicles, and ductus deferens but not the prostate. | [159] |
MMTV/wap | Develop hyperplasia/dysplasia in the anterior
gland epithelium with impaired mammary development. The transgene was expressed at elevated levels in organs with exocrine functions, including salivary and mammary glands, prostate, seminal vesicle, and coagulation gland. No invasive carcinoma, no metastasis. |
[160] |
Two major transgenic mouse lines have been created using distinct versions of the prostate-specific PB transcriptional regulatory elements. In the first mice model, the transgenic line carrying the rat PB gene was fused with the SV40 early region designated as the TRAMP (transgenic adenocarcinoma of the mouse prostate) line of mice. This model is the first autochthonous mouse model generated and characterized in 1996 [55][56]. This model exhibits various disease manifestations, and the median survival rate for these mice is 42 weeks. Most of these adenocarcinomas are confined to the DLP and VL epithelial cells, most analogous to the peripheral zone where most tumors originate in humans [57][58]. With a C57BL/6 genetic background, they histologically exhibit mild to extensive hyperplasia with cribriform structures by 8-12 weeks and later progress to carcinoma by 16-28 weeks [43]. By 24 to 30 weeks into the process, most of these mice have developed poorly differentiated carcinomas with neuroendocrine features and the ability to metastasize [37]. The castrating of TRAMP mice at 12 weeks of age showed no effect on primary tumor development or metastasis, as most (80%) of mice eventually developed poorly differentiated neuroendocrine carcinomas along with metastases by 24 weeks [60]. The incidence rate of metastasis in castrated mice was represented twice as compared to non-castrated TRAMP controls [60]. Remarkably, a mixed genetic background C57BL/6/FVBn exhibited enhanced tumor incidence in comparison to the parental strain and acquired bone metastasis at low frequency [8]. Hence, TRAMP model is considered as a reliable source of primary and metastatic tumors for in-depth analysis, aiming to elucidate the earliest molecular events in the onset, progression, and metastasis of PCa [61].
TRAMP mice model is referred to as a model to study NEPC characteristics; nevertheless, this mice model is recognized as effective for studying the mechanism involved in transforming “AR driven to non-AR driven” phenotypes of PCa [62][63][64]. In the TRAMP model, the disease advances progressively, displaying multifocal and heterogeneous characteristics [59]. Tang et al. utilized the TRAMP model to illustrate the tumor progression and the emergence of neuroendocrine carcinoma (NEC) in mice following chemotherapy (docetaxel) and/or hormone excision (castration). NEC was detected in both the castration-only and docetaxel-only treatment groups, with a notably higher incidence observed in the group receiving combined treatment (castration and docetaxel) [65]. Additionally, NEC tumors exhibited an elevated proliferative index, higher metastasis potential and drug-resistance [65]. Furthermore, Niu et al., employing prostate epithelial and stromal AR knockout or prostate epithelial-specific AR knockout TRAMP models, identified that the loss of AR in either mice led to the development of poorly differentiated primary tumors characterized by expanded intermediate cell populations [66]. In investigating the human genetic component of PCa susceptibility, a study explored how mouse strain background (FVB and B6) influences the TRAMP model. They observed that FVB-TRAMP mice had shorter lifespans due to increased neuroendocrine precursor lesions detected at 4 weeks compared to B6-TRAMP mice [67]. In addition, Gao et al., using the TRAMP model, investigated the involvement of the AKT1-β-catenin pathway in advanced PCa. The study revealed that Akt1 deficiency in mice inhibits oncogenic transformation, significantly reducing tumor size in TRAMP/Akt1+/− mice and the complete absence of tumors in TRAMP/Akt1−/− prostate. Conversely, late inhibition of Akt activity in 25-week-old tumor-bearing TRAMP mice promoted PCa metastasis to the lung [68]. Moreover, utilizing TRAMP model to examine the role of Znt7 (a Zinc transporter) revealed that Znt7-null TRAMP mice exhibited higher PIN and faster primary and metastatic prostate tumors [69]. Hence, the TRAMP mice model is considered the most suitable for investigating the potential role of gene and molecular mechanisms and studying agents that target hormone-refractory disease.
Another transgene line, the LPB-Tag (LADY) PCa mouse model, is pathologically related to the TRAMP model and was generated in 1998 [70]. Multiple LPB-Tag mouse lines have been created with a deletion in the early region to remove expression of the small Tag, ensuring consistent and elevated transgene expression in their prostates. Some contain a large fragment of PB upstream of the SV40 T-antigen, along with androgen and growth factor-responsive sequences. However, the chromosomal integration site still affects the expression level and prostate tumor growth rate. In this model, tumor growth is androgen-dependent since tumors relapse in mice castrated at 11 weeks, with no regrowth up to 40 weeks [70]. Remarkably, androgen treatment of castrated mice restores the epithelial cell versus stromal cell ratio and speeds up tumor growth. The most aggressive LPB-Tag line originated from 12T-7 and has numerous transgenes on two chromosomes. The offspring of this founder are classified into slow-growing 12T-7s and fast-growing 12T-7f lines. Both lines showed PIN with reduced invasive adenocarcinoma by 15-22 weeks. However, phenotypically, the 12T-7f line showed rapid prostate enlargement HGPINs and developed stromal hypercellularity [70]. In contrast to other mouse lines, 12T-10 has reflected slow prostate tumor growth and HGPINs without stromal hypercellularity, as seen in human HGPIN and PCa [71]. Hence, LADY mice are critical to analyzing the importance of neuroendocrine differences in the stepwise mechanism of PCa progression. The LADY model is also utilized to assess the effects of adjuvant therapy and chemotherapy agents. For instance, crossing LPB-Tag with VDR knockout mice is employed to investigate the role of VDR in prostate tumor initiation and progression [72].
The TRAMP/LADY models face limitations in oncogenesis research due to their reliance on a non-human-relevant viral oncogene for prostate-specific inactivation of pRb and p53. This leads to the development of "small cell" or NEPC tumors, which are rare in human PCa [73]. Despite these constraints, TRAMP and LPB-Tag models remain vital tools for comprehending molecular pathways in PCa and contribute significantly to preclinical chemoprevention approaches [74][75].
Table 1 describes the disease progression profile in these transgenic mice derived from the diverse promoter. Overall, T antigen-driven mouse models are widely used to study PCa biology because they can recapitulate certain characteristics of human PCa. However, these models present several drawbacks: (i) tumor development occurs at a much faster rate than typically observed in humans, limiting the study of PCa's natural history; (ii) these models often lack the histological heterogeneity seen in human PCa, a hallmark of the disease, (iii) T antigen-driven mouse models often rely on targeted genetic alterations in specific signaling pathways, restricting their ability to fully recapitulate the intricate genetic and epigenetic alterations occur in human PCa, (iv) the early expression of T antigen in these models affects prostate gland development starting from the first week of life [73][24][8][76].
4. Evolution of the Cre-loxP System: Advancements in transgenic mouse models
The Cre-loxP system was characterized by Sternberg’s laboratory from plaque-forming PI phage, which infects Escherichia coli [77]. Cre is a gene that encodes tyrosine site-specific recombinases that “causes recombination,” and loxP (locus of x-over, P1) is known as the “locus of phage crossing over.” In this way, Cre is a recombinase protein involved in site-specific genetic recombination (inversion, deletion, and translocation) in trans at loxP sites [77][12]. Cre was modified by fusing the mutant ligand binding domain of the human estrogen receptor (ER), activated by tamoxifen but not by estradiol, to accomplish more precise genetic studies and clinical applications. [78]. The benefits of conditional gene targeting are restricting Off-target effects and developmental defects. Moreover, it also allows for knocking down both alleles of interest without embryonic lethality. A strategy was employed to establish a transgenic mouse model (Figure 3A, B), resulting in the generation of various mouse lines outlined in Table 2.
Figure 3. Schematic representation of GEMM, common vectors, and mechanism of action of Cre-loxP system.
(A) Strategies for creating GEMM [16][13]: (1) blastocysts were extracted from mice, (2) embryonic stem (ES) cells were cultured, (3) a targeting vector containing DNA sequences homologous to the gene of interest is added in ES cells (4) and selected it for clonal positive selection (5) and targeted ES cells were expanded (6) Targeted ES cells were then injected into mouse blastocysts, (7) and implanted into a surrogate mother mouse, (8) developed the chimeric mouse. Screening of offspring on the basis of % presence of genetically engineered allele and this process repeated to create mice with different genetic alterations.
(B) Cre-lox system used to control site-specific recombination events in genomic DNA. When Cre recombinase encounters two LoxP sites, it catalyzes a recombination event between them. The result is a recombined locus in which the gene of interest is deleted, creating a “floxed” gene (left). For this purpose, several prostate-specific promoters have been developed, as represented (center). Two major systems, Cre-loxP and inducible Cre-loxP, were generated using these different vectors. The inducible Cre-loxP comprises Cre recombinase and a driver or promoter that controls when and where Cre is expressed, allowing for targeted gene expression manipulation (right). The principle of tamoxifen (T)-an inducible system of estrogen receptor fused to Cre (CreER) is: in the absence of T, expressed fusion protein, CreER, interacts with heat shock protein 90 (HSP90) and exists in the cytoplasm. (1) the interaction of HSP90 with CreER is disrupted after administration of T, (2) nuclear translocation of Cre happens with the interaction of ER and T, (3) the CreER recognizes the loxP sites in the nucleus, (4) and inactivates the gene X in a specific tissue.
Table 2.
Transgenic mouse lines expressing Cre recombinase/tamoxifen induced Cre recombinase in prostatic epithelial cells
Mouse Model | Recombinase | Recombinase activity |
Ectopic Cre expression |
Transcriptional regulatory elements |
References |
---|---|---|---|---|---|
Pb-Cre | Cre - recombinase | VP > DP, AP, LP | Bladder, seminal vesicles | Rat PB (−426bp to+28bp) | [161] |
Pb-Cre4 | Cre - recombinase | LP > VP, DP, AP | Seminal vesicles, seminiferous tubules, prostatic Basal and stromal cell | ARR2PB | [162] [11] |
ARR2PBi-Cre4 | Cre - recombinase | LP, VP, DP, AP | Seminal vesicles, ductus deferens | ARR2PB | [163] |
PSA-Cre | Cre - recombinase | Mature prostate lobes | - | PSA (6 kb) | [80] [79] |
PSA-Cre-ERT2 | Chimeric- Cre-recombinase; Cre-ERT2 | Luminal cells; DLP, VP> AP | - | PSA (6 kb) | [164] |
ARR2PB-Cre-ERT2 | Chimeric- Cre-recombinase; Cre-ERT2 | Luminal cells; AP, DLP> VP | - | ARR2PB | [165] |
PB-MerCreMer | Chimeric- Cre-recombinase; MerCreMer | Luminal cells; AP, DLP> VP | - | PB genomic region | [166] |
Nkx3.1Cre-ERT2 | Chimeric- Cre-recombinase; Cre-ERT2 | Luminal cells, Basal cells (10%) | - | Nkx3.1 | [85] |
The creation of the androgen-responsive ARR2PB (PB-Cre4) marks a substantial advancement in prostate-specific gene targeting, serving as a valuable tool for genetic-based PCa research [11]. Using the PSA-Cre promoter PSACre;Nkx3.1flox, and PSACre;PtenloxP/loxP mice were generated, showing PSA-Cre activity in all prostate lobes [79][80][81]. The Catnb+/Δex3; MMTVcre mice were engineered to activate the Wnt/β-catenin pathway by deleting exon 3 in the β-catenin gene across various cell types. As a result, the stabilization of β-catenin in prostate epithelium led to hyperplasia and substantial transdifferentiation in to epidermal-like structures [82]. Similarly, the PtenloxP/loxP;MMTV-cre mice were developed to selectively inactivate Pten in mouse tissue, resulting in the development of HGPIN that often advanced to focal invasive PCa [83]. However, these models have also been reported to develop off-target effects. FSP1(Fibroblast-specific protein 1) -Cre-driven gene deletion is reflected as a reliable strategy to explore fibroblast biology. The FSP1-Cre TGFflox (Tgfbr2fspKO) mice model was generated to understand the role of transforming growth factor-beta (TGF-β) pathway and epithelial-mesenchymal connections by conditional inactivating the TGF- β type II receptor gene in fibroblasts [84]. The limitation of this promoter is gene recombination occurs in many tissues due to the universal presence of fibroblast throughout the body. Therefore, this model can be utilized to recognize the role of TGFβ in tissue differentiation but not a specific study for PCa. Next, tamoxifen induced Cre activity used to generate NKx3.1CreERT2/+; R26RYFP mice. Through genetic lineage-marking, it has been revealed that there are rare luminal cells within the prostate known as castration-resistant Nkx3.1-expressing cells (CARNs) exhibit bipotential properties and have self-renewal potential in vivo. Wang et al. demonstrated that these cells exhibit stem/progenitor properties within the androgen-deprived prostate epithelium during prostate regeneration, suggesting that Nkx3-1 is needed for stem cell maintenance [85]. In addition the targeted deletion of the Pten in CARNs (Nkx3.1CreERT2/+;Ptenflox/flox mice), results in rapid onset of carcinoma subsequent to androgen-mediated regeneration [85]. Hence, this model has clinical relevance and could be targeted for CRPC.
5. Second generation genetically engineered mouse models of PCa: Integration of Human genomic alterations
GEMMs are sophisticated tools that have evolved from the single allelic transgenic model to generate complex multiallelic genetic alterations that more accurately mimic the genomic and molecular changes observed in human cancers, including PCa [9]. Table 3 presents an overview of GEMMs designed to mimic different stages of PCa and based on human genomic alteration. Table 4 represents the mice models based on AR, WNT, TGFβ, RAS/RAF/MAPK signaling dysregulation and epigenetic modifications.
Table 3.
GEMM based on human genomic alteration using different promoters.
Mouse Model | Features | Ref. |
---|---|---|
Pten-derived mouse
model with multiple genetic mutations |
Loss of function | |
Pten+/− | Develop dysplastic intestinal polyps, PIN, and
neoplasia in multiple tissues. Most die within 8 months due to massive lymph-splenomegaly. The survivor PTEN+/− mice display PIN lesions by 10 months. No invasive and metastatic adenocarcinoma. |
[89][88] |
Ptenflox/flox(exon 4,5)/PBCre4 | Develop HGPIN in 9 weeks, followed by
carcinoma in 17-26 weeks. No metastasis up to 130 weeks. |
[90] |
Ptenflox/flox(exon 5)/PBCre4 | Develop prostatic epithelial hyperplasia by 4
weeks, PIN in 6 weeks, and adenocarcinoma by 9-29
weeks. Metastasize to lung and lymph nodes by 12 weeks. |
[95] |
Pten+/−;p27−/− | Develop PCa at complete penetrance within 12
weeks from birth. No metastasis |
[167] |
Pten+/−; Nkx3.1−/− | Develop HGPIN by 6 months of age and ultimately progress to adenocarcinoma. | [117] |
Pten+/−; Nkx3.1+/−p27+/− or Pten+/−; Nkx3.1+/−p27−/− | PCa progression is enhanced by the loss of one wildtype p27kip1 allele but inhibited by the loss of both alleles. | [97] |
NPKEYFP mice (Nkx3.1CreERT2/+; Ptenflox/flox; KrasLSL-G12D/+; R26R-CAG LSL-EYFP/+ | Tumor formation at 2- 3 months of age by tamoxifen administration and micro-metastasis at 5-8 months. | [102] |
Pten−/+; Trp53−/− (RapidCap- model) | Metastasis to distant sites at greater than 50% penetrance by four months. | [105] |
NPp53 mice (Ptenflox/flox; Trp53flox/flox; Nkx3.1CreERT2) | Under castrate, develops CRPC characteristics and, after treatment with abiraterone, displays NEPC features. | [109] |
Ptenflox/flox: Rbflox/flox/ PBCre4 (DKO model) | Develops PIN and early invasive carcinoma
within 12 weeks. Neuroendocrine features (synaptophysin-positive cells) appear by 20-25 weeks. ADT sensitive because castration extended the median survival of these mice from 38 to 48 weeks, but all mice eventually died from prostate cancer by 67 weeks. Median survival 38 weeks. Develops metastasis in the lymph nodes, liver, and lung. |
[122] |
Ptenflox/flox;Rbflox/flox;Trpflox/flox/ PBCre4 (TKO model) | Develops castration-resistant tumors, survival
rate 16 weeks. ADT-resistance because castration of these mice at 10 weeks did not extend survival. These TKO tumors have reduced dependence on both AKT and AR signaling. |
[122] |
Hi-Myc-derived mouse
model
with multiple genetic alterations |
Conditional Gain of function | |
Hi-Myc/ARR2PB; Hepsin | Invasive adenocarcinoma at 4.5
months. This model develops a higher-grade adenocarcinoma in 12- to 17-month-old mice. Tumors showed higher hepsin expression. The proposed mechanism is basement membrane degradation. No metastasis. |
[168] |
Hi-Myc/ARR2PB; IκBa+/− | Develops adenocarcinoma at 26
weeks. PCa progression to CRPC. The proposed mechanism is the activation of NF-κB signaling by regulating AR action. |
[169] |
Z-Myc-derived mouse
model
with multiple genetic alterations |
Conditional loss of function | |
Z-Myc/PBcre4; PtenFl/Fl | HGPIN/cancer lesions and promote faster
prostate tumorigenesis. The proposed mechanism is a bypass from senescence to apoptosis by repressing the p53 target gene p21Cip1. |
[170] |
Z-Myc/CMV-β-actin; Nkx3.1 | HGPIN with microinvasion. MYC knockdown dysregulates shared NKX3.1/MYC target gene expression. The enhanced MYC transcriptional activity. |
[171] |
Z-Myc/PBcre4; Pten+/−;TrP53−/− | PIN and adenocarcinoma (10-24 weeks) with
marked heterogeneity within the same prostate glands. It accelerates tumor formation and leads to lymph node metastases. |
[172] |
BMPC model (Hoxb13-Myc;Hoxb13-Cre PtenFl/Fl) | Develops lethal adenocarcinoma with distant
metastases. BMPC cancers are deficient in neuroendocrine/ or sarcomatoid differentiation. Induce genomic instability and aggressive prostate cancer similar to human disease at the histologic and genomic levels. |
[98] |
Other models | Conditional modulation of gene expression | |
Trp53−/−; Rb−/− | PIN lesions at 8 weeks. Poorly differentiated neuroendocrine features by ~32 week Tumors are ADT-resistant de novo because castration does not extend survival. |
[73] |
Trp53R270H/+ Nkx3.1Cre | PIN as early as 5 weeks, well-developed
neoplastic foci as early as 4 months. Displays invasive adenocarcinoma with epithelial-mesenchymal transition (EMT) phenotype. |
[108] |
Tmprss2-Erg fusion/Pten−/+ | Acceleration of HGPIN and progression to
prostatic adenocarcinoma at 6 months. The proposed mechanism is by modulating AR signaling and checkpoint genes. |
[135] |
Spop/Pten−/+ | Develops early neoplastic lesions (HGPIN) with striking nuclear atypia and invasive poorly differentiated carcinoma. Activates both PI3K/mTOR and AR signaling. | [138] |
PBCre4/Foxa1loxp/loxp | Develops hyperplasia with an extensive
cribriform pattern but does not expand toward more advanced
phenotypes. Alterations in localization of p63 positive basal cells. Following castration, the conditional knockout Foxa1 retained its androgen sensitivity, and Foxa1-positive cells appeared to be more castration sensitive. |
[173] |
Table 4.
GEMMs through alteration of molecular signaling pathways:
Molecular signaling pathways |
Features | Reference |
---|---|---|
AR | AR transgene expression in the prostate epithelium develops PIN. | [174] |
The mutant AR (AR-E231G) expressed under the control of the PB promoter facilitates the PIN. This condition subsequently progressed to invasive and metastatic disease. | [175] | |
The PB-driven ARv567es transgenic mouse model displays epithelial hyperplasia at 16 weeks and develops invasive adenocarcinoma by 1 year of age. | [176] | |
The transgenic line (R26hAR(loxP): Osr1-Cre+) exhibits conditionally activated AR. Transgene expression is seen in both prostatic luminal and basal epithelial cells and is resistant to castration. | [177] | |
Wnt/ β-catenin | Conditional loss of function of adenomatous polyposis coli (Apc) develops HGPIN and adenocarcinoma depending on Cre-driver. | [178] |
Conditional activation of β-catenin represents PIN phenotypes, and cancer progression is achieved by collaboration with the loss of function of Pten | [179] [180] [181] |
|
Synergism of activated K-ras (K-ras+/V12) and dominant stabilized β-catenin (Catnb+/lox(ex3)) with PbCre promoter induces rapid progression of prostate tumorigenesis to invasive carcinoma. | [182] | |
sδ-catenin (gene designation: CTNND2) mutations promote tumor development in mouse prostate with ARR2PB promoter | [183] | |
Combining Wnt-signaling and Tag expression in the mouse prostate (LPB-Tag/dominant active mice) display neuroendocrine differentiation (NED), but NE cancer does not develop. | [184] | |
TGF- β | Model in PCa to study stromal interaction. | [84] |
PbCre4 promoter-dependent conditional loss of Smad4 used for TGF-β activation. Combined conditional loss of Pten and Smad4 develop aggressive adenocarcinoma, characterized by a short latency period and distant metastasis with low penetrance. | [100] | |
In addition, conditional loss of Pten, Smad4 and Trp53, along with telomerase deficiency, represent invasive adenocarcinoma with extensive metastasis (bone metastasis). | [110] | |
COUP-TFII (NR2F2) inhibits Smad4-dependent transcription, and overexpression of COUP-TFII in the mouse prostate epithelium cooperates with Pten deletion develops an aggressive metastasis-prone tumour. | [185] | |
Ras/Raf/MAPK | Conditional gain of expression of mutated gene Kras G12D with Pten-based background in the prostate of mice represents invasive adenocarcinoma with and macrometastasis with 100% penetrance. | [186] |
An activating mutation of BrafV600E induced GEM to develop prostate gland hyperplasia with rapidly growing invasive adenocarcinoma. | [187] | |
Conditional gain of expression of NCoA2 (nuclear receptor coactivator 2) with Pten loss of function background shows invasive adenocarcinoma with distant metastasis. | [188] | |
Epigenetic regulator | Histone methyltransferase WHSC1 overexpression in prostate epithelium, combined with Pten deletion, resulted in a metastasis-prone tumor in a Pten-null murine PCa model. | [189] |
5.1. GEMMs based on tumor suppressor genes
5.1.1. Pten-derived mouse model with multiple genetic mutations
PTEN (phosphatase and tensin homolog deleted from chromosome 10), known as Mmac1, is a crucial tumor suppressor gene, and its loss of function is associated with ~ 35% of primary PCa and ~63% of advanced/ metastasis [86][87]. The first Pten knockout mouse was developed by creating a null mutation in 1998; one more group in 1999 also created a Pten knockout mouse by deleting the distinct portions of the Pten gene that showed similar inactivating activity [88][89]. These null models were embryonically lethal, and no homozygous knockouts were alive [89]. The various models of Pten-based GEMM have been developed, which differ based on the severity of disease phenotypes, prevalence, latency, and biology. The differences in severity depend on the timing of gene recombination comparative to prostate differentiation, contextually to specific cell types targeted for recombination, selection of precise Cre-driver, specific deleted region of Pten exon, mouse strain background, and variance in phenotypic-pathological interpretation [90][91]. To assess the impact of varying PTEN levels on cancer progression through homologous recombination, a series of hypomorphic Pten mouse mutants ( Ptenhy/+, Pten+/−, and Ptenhy/−) with decreasing Pten activity was created [90]. It has been observed that the Pten dosage dictates the PCa progression and regulates critical downstream targets, including Akt, p27(Kip1), mTOR, and FOXO3 [90]. A recent study highlighted the importance of the dynamic interplay between mTOR and AMPK activities in metastatic colonization by circulating PCa cells, emphasizing the concurrent targeting of both AMPK and mTOR [92]. Bertram et al. reported that Pten inactivation is adequate for acquiring androgen independence progression [93]. A recent study investigated the impact of age on PCa using a controlled Pten-null mouse model; both aged and non-aged adult mice developed prostatic epithelial hyperplasia within 4 weeks, PIN within 8 weeks, and some PINs progressed to invasive adenocarcinoma between 8 to 16 weeks post-Pten ablation. Notably, aged mice showed accelerated PI3K/AKT/mTOR signaling and increased onset and progression of PCa compared to young mice [94].
The conditional Pten deletion in the prostatic epithelium showed progression towards hyperplasia after 4 weeks, PIN in 6 weeks, and full adenocarcinoma (100% penetrance) at 9-29 weeks [95]. Histopathologically, the Pten null tumor originated from secretive epithelial, a source similar to most human prostate tumors [95]. Notably, the invasive Pten-null PCa cells did respond to androgen ablation and showed increased apoptosis, mirroring human PCa. Even though the survival of these cells was androgen-sensitive, their proliferation remained unaffected. A global evaluation of molecular changes in Pten-null mice (homozygous deletion) identified the “signature” gene expression changes similar to those relevant for human PCa and other human cancers with metastatic potential. This model provided an exclusive tool for investigating the molecular process of PCa metastasis and developing new targeted therapy [95]. The Pten mouse model has some limitations in its histological representation, such as the development of squamous differentiation, sarcomatoid carcinoma, and mucinous metaplasia in some instances [96].
Interestingly, triple mutant mice (Nkx3.1+/− or −/−; Pten+/−; p27+/−) that were heterozygous for a p27kip1 null allele exhibited accelerated prostate carcinogenesis, which is contrary to the findings observed in homozygous null for p27Kip1 (Nkx3.1+/− or −/−; Pten+/−; p27−/−) mice, which showed suppression of PCa progression [97]. Expression profiling revealed that compound p27kip1 heterozygotes exhibited higher Cyclin D1 levels, while these levels were downregulated in compound p27kip1 homozygous mutants [97]. The loss of Pten with other regulatory proteins has also been investigated; all contribute to PCa progression and metastatic phenotypes. These includes MYC activation (Hoxb13-MYC;Hoxb13-Cre;PtenFl/Fl), participation of ETV4 via PI3K and RAS signaling and the functional significance of SMAD4 via TGFβ/BMP–SMAD4 signaling (Ptenloxp/loxp;Smad4loxp/loxp;PB-Cre4) [98][99][100]. Furthermore, a study indicated that the transgenic expression of a constitutively activated p110β (Pi3kcb) allele in the prostate leads to PIN formation. The resulting lesions resemble those caused by PTEN loss or Akt activation but exhibit clear distinctions [101].
Recently, a new NPKEYFP mice model (Nkx3.1CreERT2/+; Ptenflox/flox; KrasLSL-G12D/+; R26R-CAG LSL-EYFP/+) was developed for detailed biological and molecular characterization of lethal PCa related to bone metastasis and castration-resistance [102]. These mice use the inducible Cre guided by NKX3.1 promoter to accomplish spatiotemporal regulation of gene recombination (Ptenflox/flox; KrasLSL-G12D/+), particularly in prostatic luminal cells. Visualization of tumor and bone metastasis is made possible by activating the fluorescent reporter allele (R26R-CAG LSL-EYFP/+). Through tamoxifen administration, NPKEYFP mice were induced to tumor formation at 2-3 months and monitored for micro-metastasis at 5-8 months. This model developed PCa with high penetrance for metastasis and is capable of homing to the bone. The median survival of NPKEYFP mice (4.7 months) was significantly higher than control NPK (3.1 months) mice. Sequencing data analysis revealed a link between MYC/RAS co-activation and PCa metastasis, underscoring the importance of the META-16 gene profile in adverse outcomes and treatment response [102].
5.1.2. TrP53-derived mouse model with multiple genetic alterations
TRP53 mutated or deleted in >50% of aggressive PCa [103]. The alteration of P53 expression either by losing one copy of the Trp53 gene or by a mutation in transgenic mice PCa model presented HGPIN lesions (grades III-IV) by 52 weeks with decreased apoptotic rate [104]. The germline or conditional loss of function of Trp53 based GEMMs model represented modest PCa phenotypes, with only PIN lesions not transformed towards adenocarcinoma.
To evaluate the therapeutic potential for metastatic PCa, the RapidCaP mouse model was designed to utilize a surgical technique to introduce lentiviral transgenes into the prostate. This model is established with the combined loss of function of Trp53 and Pten genes and displays metastasis to distant sites (> 50% penetrance) within 4 months [105]. Prostate-specific deletion of Pten and Trp53 by incorporating a Cre-activatable luciferase reporter resulted in uniformly lethal disease by 25 weeks [106]. The functional consequence of Trp53 alteration in conjunction with Pten loss in the prostate epithelium is cell lineage plasticity [107]. Another study revealed that prostate-specific Trp53 mutant Trp53R270H expression, i.e., similar to the human hotspot mutant R273H, indicated a moderate PIN phenotype [108]. This feature was augmented with a combination of loss of function of Nkx3.1+/−. Furthermore, the related loss of Trp53 and Pten with Nkx3.1, Pten flox/flox; Trp53flox/flox; Nkx3.1CreERT2 (NPp53 mice) following castration encouraged CRPC with neuroendocrine differentiation phenotypes [109]. These NPp53 mice are highly aggressive and resistant to anti-androgen treatment, and notably, anti-androgen therapeutic approaches aggravate disease progression [109]. Preclinical analyses revealed that abiraterone treatment caused modest but significant suppression of NP CRPC. At the same time, NPp53 CRPC models were resistant to abiraterone, as apparent from their histopathology and absence of reduced cellular proliferation or tumor volume. The NPp53 mouse model displays accelerated tumor progression, with features similar to treatment-related CRPC with neuroendocrine differentiation as seen in humans. Therefore, these models are not the best choice for studying the efficacy of abiraterone or other AR-targeted therapies in CRPC [109]. Moreover, it has been speculated that telomerase reactivation following telomere dysfunction generates prostate tumors with bone metastases. Study reported that the inducible telomerase reverse transcriptase (mTert) allele crossed over with Pten null and Trp53 developed aggressive cancers with rearranged genomes and new tumor biological features [110].
5.1.3. Nkx3.1-derived mouse model with multiple genetic mutations
The allelic deletion of NKX3.1 has been reported in ~80% of prostatic neoplasia and is considered a gatekeeper suppressor gene [111][112]. The first Nkx3.1 knockout mouse was established in 1999 to examine its physiological effects, and numerous phenotypical changes were observed in the prostate and seminal vesicles [113]. Loss of Nkx3.1 (conventional and conditional) promoted initiation of dysplasia and hyperplasia in an age-dependent manner, but no overt tumors were observed [79][112]. The targeted interruption of Nkx3.1 in mice showed morphogenetic defects in the salivary gland, suggesting its role in developing other cell types [114]. Bowen et. al investigated that the absence of NKX3.1 expression is highly correlated with hormone-refractory disease and advanced tumor stage in PCa [115]. In another study, aged Nkx3.1 mutant mice exhibited histopathological abnormalities resembling PIN and suggested that the loss of Nkx3.1 function is a pivotal event in the initiation of prostate cancer and serves as a model for early-stage disease [116].
Notably, Nkx3.1; Pten double mutant mice showed an augmented incidence of HGPIN, invasive adenocarcinoma, and lymph node metastasis, highly similar to the early stage of human PCa [117]. Loss of Nkx3.1 coordinates with Pten loss for PCa progression, and this cooperativity is mediated by synergistic activation of AKT /protein kinase B [117]. Another finding affirmed the function of PTEN as a phosphatase for NKX3.1, opposing phosphorylation at NKX3.1(S185) to preserve its stability and extend half-life. In gene-targeted mice, Pten loss significantly reduced Nkx3.1 expression, promoting prostate epithelial cell proliferation [118].
5.1.4. Rb-derived mouse model with multiple genetic alterations
Retinoblastoma (RB), a tumor suppressor gene at chromosomal loci 13q, exhibits a specific mutation linked to early events in PCa [119][120]. Functional RB family proteins (Rb/p107/p130) inactivation via conditional deletion of the RB gene in prostate epithelium stimulates epithelial proliferation and apoptosis. This model produced PIN and adenocarcinomas with no indication of neuroendocrine tumors [120]. The conditional loss of Rb function in the prostate reflected a relatively modest phenotype, but collaboration with other genomic alterations accelerated PCa progression [121][73]. In particular, a Trp53PE−/−: RbPE−/− knockout mice model developed PIN lesions as early as 8 weeks and a poorly differentiated phenotype with neuroendocrine features by 32 weeks. Rb and TrP53 loss was observed to suppress the epigenetic reprogramming factors (EZH2 and SOX2), creating a stem-cell-like epigenetic environment permissive to lineage plasticity [73].
Next, DKO mice (PBCre4: Ptenf/f: Rbf/f) had short latency, established PIN lesions by 12 weeks, had distant metastasis, and displayed shorter survival (38 weeks) than single gene mutant models (48 weeks). In DKO mice, Rb loss acted as an enhancer of lineage plasticity stimulated by Pten loss. The DKO model, sensitive to ADT, exhibits reduced AR expression, spontaneous Trp53 mutations (V173M, R282Q), and tumors relapsed following castration [122]. TKO mice (PBCre4: Ptenf/f: Rbf/f: Trp53 f/f) had a concise survival rate of 16 weeks and reflected aggressive phenotypes. Despite androgen deprivation, tumor biology, and survival were unaffected, confirming the insensitivity of these tumors to castration. This model unveiled that Rb1 loss facilitates lineage plasticity and metastasis initiated by Pten mutation, while additional Trp53 loss results in resistance to antiandrogen therapy [122].
5.2. GEMMs based on activation of gene
5.2.1. Myc-derived mouse model
The MYC overexpression in the prostate depends on the level of MYC expression as high (Hi) or low (Lo) GEMMs have been developed [123]. The PB/ Lo-Myc model comprised minimal PB promoter expressed a low level of MYC, and PIN lesions were perceived at 10 weeks, showing progressive to invasive adenocarcinoma by 10-12 months. In comparison, the ARR2PB / Hi-Myc model consists of a reconstructed PB promoter expressing a high level of MYC, and the PIN lesions were noticed at 2 weeks, showing prostate intraepithelial neoplasia to invasive adenocarcinoma by 3-6 months. Although PB/ Lo-Myc and ARR2PB / Hi-Myc mice showed similar phenotypes at pathological terms, the difference is only the duration of pathological changes and their androgen responsiveness [123][124]. Furthermore, Hi/Myc mice progressed towards CRPC following androgen deprivation [123]. A characteristic feature of Myc-based models is that they do not exhibit sarcomatoid or neuroendocrine differentiation histopathology [98]. Hence, this model is pronounced as an accurate representative model of human adenocarcinoma. Moreover, several Myc based GEMM cooperativity with other genes have been developed, as discussed in Table 3.
Particularly, the BMPC model (Hoxb13-Myc; Hoxb13-Cre; Ptenflox/flox) had a higher level of MYC and tended to develop invasive carcinoma with distant metastasis to lung, liver, and bone rarely. Tumors of BMPC mice displayed low AR and NKX3.1 but did not exhibit sarcomatoid characteristics prevalent in other advanced PCa GEMMs [98]. Dysregulation of N-MYC cooperating with myristoylated AKT1 led to the development of adenocarcinoma and NEPC, presenting phenotypic and molecular traits attributed to aggressive, late-stage human PCa [125]. Pharmacologic disruption of N-MYC expression through Aurora A kinase inhibition (AURKA inhibitor CD532) reduced the tumor burden [125].
The GEMM (T2-Cre+/+; Ptenf/+; LSL-MycN+/+) carried a CAG-driven lox-stop-lox human N-Myc gene incorporated into the ROSA26 (LSL-N-Myc) locus and a Tmprss2-driven tamoxifen-activated Cre recombinase (T2-Cre) that facilitates its expression in luminal prostate cells [126]. This model has also been engineered to harbor a Pten conditional knockout allele. In this model, tamoxifen-induced Cre activation resulted in N-MYC overexpression in the prostate, persisting from 5 weeks to 3 months post-induction, leading to HGPIN at 3 and 6 months. Subsequently, a substantial, invasive prostate tumor emerged at 9 months, featuring AR-positive adenocarcinoma, poorly differentiated carcinoma foci, and divergent differentiation [126].
5.2.2. MPAKT transgenic model
The tumor phenotype in Pten−/−embryonic stem cells depends on AKT activation [127]. The MPAKT (murine prostate-restricted AKT kinase transgenic mice) model is generated by introducing a plasmid insert containing a PB promoter, a myristoylated sequence (myr), and a hemagglutinin (HA) epitope-tagged sequence, and human AKT1in the form of rPB-myr-HA-AKT1 insert into fertilized oocytes pronuclei. This resulted in the expression of the myristoylated AKT1 oncogene, specifically in the prostate epithelium. The MPAKT mice developed PIN lesions in the ventral prostate, accompanied by significant bladder obstruction, but without any signs of metastasis [128]. At the molecular level, the MPAKT mice represented activated AKT1 spatially confined to the prostate and showed activation of the p70S6K pathway. This model is valuable for examining the role of AKT/ PKB in prostate epithelial cell transformation and discovering biomarkers relevant to human PCa [95].
5.3. GEMMs based on genetic rearrangements, mutation, and alteration:
5.3.1. TMPRSS2-ERG fusion gene mouse model:
E26 transformation-specific (ETS) gene fusion and overexpression of ETS factors ERG, FLI1, ETV1, ETV4, and ETV5 have been reported in PCa [129]. Activation of ERG is associated with PCa progression in both early- and late stages [130][131]. Expression of the TMPRSS2:ERG gene fusion regulates bone markers and promotes the osteoblastic phenotype of PCa bone metastases [132]. Inducing targeted expression of the ERG in luminal prostate epithelial cells of transgenic mice reflected the initiation of prostate neoplasia, characterized by the development of focal precancerous PIN. Notably, like human cancers, luminal epithelial cells in these PIN lesions reduce the number of basal epithelial cells and make direct contact with the stromal cell compartment. Hence, this study indicates that ERG plays a vital role in the transformation of prostate epithelium and is considered a promising target for early therapeutic intervention [133]. Notably, PB-driven ERG, ARR2PB-driven ETV1, or ERG over-expression resulted in only PIN and no other phenotypes over the mouse lifetime [134][135]. In addition, ARR2PB-driven TMPRSS2-ERG exhibited no histological phenotype for up to 60 weeks [136]. The concomitant TMPRSS2-ERG expression and PTEN heterozygous background transgenic mice promoted the marked acceleration of HGPIN and invasive prostatic adenocarcinoma at 6 months of age [135].
5.3.2. SPOP and other early alteration-related mouse models:
Genomic instability/ impaired genome maintenance resulting from point mutations in E3 ubiquitin ligase SPOP (Speckle-type POZ protein) is a typical mechanism driving prostate tumorigenesis and occurs in ~10% of PCa [137]. SPOP-mutant PCa represents genetically and biologically distinct subsets that respond to AR signaling inhibition [7]. A GEMM with prostate-specific conditional expression of mutant Spop (SpopF133V) showed a modest phenotype. The setting of Pten loss with Spop mutation reflected dramatically altered phenotypes such as early neoplastic lesions (HGPIN) with dramatic nuclear atypia and invasive poorly differentiated carcinoma [138].
The SPOP mutations and CHD1 (chromatin remodeler chromodomain helicase DNA-binding protein 1) homozygous deletions frequently co-occur in PCa patients [139][140]. The GEMM PB-Cre+; Chd1L/L mice led to the development of PIN but did not develop PCa. Mechanistically, this model showed changes in DDR (DNA damage repair) by modulating the choice between HR (homologous recombination) and NHEJ (non-homologous end joining) DNA double-strand breaks (DSBs) repair. Remarkably, loss of CHD1 reduces HR-mediated DSB repair and enhances error-prone NHEJ activity [139]. FOXA1 (forkhead box) alteration in PCa was mutually exclusive with the alteration of SPOP and ETS fusion [141]. The impact of prostate-specific FOXA1 deletion in the epithelium of adult mice indicated progressive florid hyperplasia with an extensive cribriform pattern but did not progress toward more advanced phenotypes [142].
Conclusion & Future Insights
Overall, mouse models have proven valuable tools for PCa research but have several limitations, as discussed. An ideal mouse model should adhere to the following principles: (i) it should encompass all stages of PCa initiation and progression, exhibiting easily identifiable features that mirror both the histopathological and molecular aspects of human PCa; (ii) at early stages, the model should demonstrate AR biology and exhibit a slow progression rate; additionally, it should possess the ability to metastasize to the same sites as human PCa, while maintaining an intact immune system. The emerging concept of personalized mouse models, Co-clinical Trials, has been introduced to overcome these pitfalls. These trials enable the real-time integration of the murine and human tumor data. In Co-clinical Trials, GEMMs are utilized to guide therapy in an ongoing human patient trial toward future clinical management of the patient's tumor [143].
GEMMs are considered indispensable for preclinical research and superior to cancer cell inoculation models. One unique feature of GEMMs is the ability to develop de novo tumors in a natural immune-proficient microenvironment. In addition, advanced GEMM-derived tumors closely resemble the histopathological and molecular characteristics of their human counterparts, exhibiting genetic diversity and progressing spontaneously toward metastatic conditions [144]. Furthermore, GEMMs of PCa represent potent tools for investigating the mechanisms underlying disease progression and the response to therapy. A recent study detailed the development, examination, and validation of a platform designed to immobilize and precisely target tumors in mice using stereotactic ablative radiation therapy (SART). This approach has been observed to modify the tumor stroma and immune environment, improving survival outcomes in GEMMs of primary PCa and exhibiting synergy with androgen deprivation [145].
Nevertheless, despite the SART application, complete pathologic responses were not attained in GEMMs. Consequently, investigating the mechanisms of radiation resistance in GEMMs of PCa presents an intriguing area for further exploration. The onset, advancement, and development of CRPC entail intricate interactions between tumor cells and the host immune system [146]. GEMMs offer a unique prospect to investigate the inherent factors influencing tumor response and assess the impact of extrinsic determinants. It has been reported that GEMMs of PCa have differing quantities of tumor-infiltrating lymphocytes characterized by notable spatial heterogeneity [145].
Using these mouse models has led to the development of new imaging techniques that can detect bone metastases at an earlier stage and tumor microenvironment mechanisms for bone metastatic disease progression, potentially allowing for more effective treatment [102][147]. Human PCa represents a heterogeneous diversity at inter- and intra-tumor and inter-patient levels, which significantly influences the prognosis, choice of therapy, recurrence, and emergence of treatment resistance in PCa [148]. In treatment-naive primary tumors, there is pre-existing AR heterogeneity, which is magnified during AR signaling inhibitors (ARSIs) treatment and notably pronounced in CRPC [149]. After extensive research spanning several decades to create GEMMs with molecular changes akin to human PCa, no mouse model has fully reflected the heterogeneity of human tumors or accurately recapitulated the observed phenotype in humans due to genetic, evolutionary, species-specific, and clinical heterogeneity [150][151][152].
Initially, basal cells were identified as the originating cells for PCa, but emerging evidence indicates that luminal cells are now considered the preferred cell of origin for PCa [153]. The emergence of single-cell RNA-sequencing technology has offered new insights into prostate epithelium heterogeneity, which is conserved between mouse and human [154]. Therefore, basal and luminal specific promoters can be valuable in identifying drivers of PCa progression precisely to each compartment.
Nevertheless, many genes remain untested, and ongoing research is necessary to uncover new genes and improve the complexity of mouse models to reflect human PCa better. The field of GEMMs for PCa is entering a new phase known as next-generation GEMMs, characterized by applying novel tools and ideas to enhance our understanding of lethal disease biology. These advancements have the potential to lead to the foundation for more accurate and relevant models for studying PCa and developing new therapeutics.
Acknowledgments
The figures were created with the help of BioRender.com.
Funding sources
This work and the authors are, in part, supported by grants from the U.S. Department of Defense (DOD) through the Prostate Cancer Research Program under Award No (DOD) W81XWH-21-1-0640 (JAS), DOD (W81XWH2110340) (JBK), and DOD W81XWH-18-1-0308 and National Institutes of Health (NIH) U01 CA185148 (SKB).
Footnotes
Competing interests
Surinder K Batra is a founder of Sanguine Diagnostics and Therapeutics, Inc. Other authors have no competing interests to declare.
1. References
- [1].Prostate Cancer — Cancer Stat Facts, (n.d.). https://seer.cancer.gov/statfacts/html/prost.html (accessed February 25, 2023).
- [2].Berenguer CV, Pereira F, Câmara JS, Pereira JAM, Berenguer CV, Pereira F, Câmara JS, Pereira JAM, Underlying Features of Prostate Cancer—Statistics, Risk Factors, and Emerging Methods for Its Diagnosis, Curr. Oncol 2023, Vol. 30, Pages 2300–2321. 30 (2023) 2300–2321. 10.3390/CURRONCOL30020178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Würnschimmel C, Wenzel M, Wang N, Tian Z, Karakiewicz PI, Graefen M, Huland H, Tilki D, Radical prostatectomy for localized prostate cancer: 20-year oncological outcomes from a German high-volume center, Urol. Oncol. Semin. Orig. Investig 39 (2021) 830.e17–830.e26. 10.1016/J.UROLONC.2021.04.031. [DOI] [PubMed] [Google Scholar]
- [4].Shore ND, Current and Future Management of Locally Advanced and Metastatic Prostate Cancer, Rev. Urol 22 (2020) 110. /pmc/articles/PMC7672503/ (accessed December 6, 2023). [PMC free article] [PubMed] [Google Scholar]
- [5].Ruiz de Porras V, Font A, Aytes A, Chemotherapy in metastatic castration-resistant prostate cancer: Current scenario and future perspectives, Cancer Lett. 523 (2021) 162–169. 10.1016/J.CANLET.2021.08.033. [DOI] [PubMed] [Google Scholar]
- [6].Yamada Y, Beltran H, The treatment landscape of metastatic prostate cancer, Cancer Lett. 519 (2021) 20–29. 10.1016/J.CANLET.2021.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Bernasocchi T, Theurillat JPP, SPOP-mutant prostate cancer: Translating fundamental biology into patient care, Cancer Lett. 529 (2022) 11–18. 10.1016/J.CANLET.2021.12.024. [DOI] [PubMed] [Google Scholar]
- [8].Parisotto M, Metzger D, Genetically engineered mouse models of prostate cancer, Mol. Oncol 7 (2013) 190. 10.1016/J.MOLONC.2013.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Arriaga JM, Abate-Shen C, Genetically Engineered Mouse Models of Prostate Cancer in the Postgenomic Era, Cold Spring Harb. Perspect. Med 9 (2019). 10.1101/CSHPERSPECT.A030528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Hanahan D, Wagner EF, Palmiter RD, The origins of oncomice: a history of the first transgenic mice genetically engineered to develop cancer, Genes Dev. 21 (2007) 2258–2270. 10.1101/GAD.1583307. [DOI] [PubMed] [Google Scholar]
- [11].Wu X, Wu J, Huang J, Powell WC, Zhang JF, Matusik RJ, Sangiorgi FO, Maxson RE, Sucov HM, Roy-Burman P, Generation of a prostate epithelial cell-specific Cre transgenic mouse model for tissue-specific gene ablation, Mech. Dev 101 (2001) 61–69. 10.1016/S0925-4773(00)00551-7. [DOI] [PubMed] [Google Scholar]
- [12].Kim H, Kim M, Im S-K, Fang S, Mouse Cre-LoxP system: general principles to determine tissue-specific roles of target genes, Lab. Anim. Res 34 (2018) 147. 10.5625/LAR.2018.34.4.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Tratar UL, Horvat S, Cemazar M, Transgenic mouse models in cancer research, Front. Oncol 8 (2018) 387140. 10.3389/FONC.2018.00268/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Doyle A, McGarry MP, Lee NA, Lee JJ, The construction of transgenic and gene knockout/knockin mouse models of human disease, Transgenic Res. 21 (2012) 327–349. 10.1007/S11248-011-9537-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Wadhwa N, Ganguli N, Majumdar SS, Generating Transgenic Animal Models: Recent Technological Advancements, Essentials Lab. Anim. Sci. Princ. Pract (2021) 709–724. 10.1007/978-981-16-0987-9_29. [DOI] [Google Scholar]
- [16].Huijbers IJ, Del Bravo J, Bin Ali R, Pritchard C, Braumuller TM, Van Miltenburg MH, Henneman L, Michalak EM, Berns A, Jonkers J, Using the GEMM-ESC strategy to study gene function in mouse models, Nat. Protoc 2015 1011. 10 (2015) 1755–1785. 10.1038/nprot.2015.114. [DOI] [PubMed] [Google Scholar]
- [17].Kumar TR, Larson M, Wang H, McDermott J, Bronshteyn I, Transgenic Mouse Technology: Principles and Methods, Methods Mol. Biol 590 (2009) 335. 10.1007/978-1-60327-378-7_22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Lee KF, Demayo FJ, Atiee SH, Rosen JM, Tissue-specific expression of the rat beta-casein gene in transgenic mice, Nucleic Acids Res. 16 (1988) 1027–1041. 10.1093/NAR/16.3.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Ivics Z, Hiripi L, Hoffmann OI, Mátés L, Yau TY, Bashir S, Zidek V, Landa V, Geurts A, Pravenec M, Rülicke T, Bösze Z, Izsvák Z, Germline transgenesis in rabbits by pronuclear microinjection of Sleeping Beauty transposons, Nat. Protoc 9 (2014) 794–809. 10.1038/NPROT.2014.009. [DOI] [PubMed] [Google Scholar]
- [20].Sander JD, Dahlborg EJ, Goodwin MJ, Cade L, Zhang F, Cifuentes D, Curtin SJ, Blackburn JS, Thibodeau-Beganny S, Qi Y, Pierick CJ, Hoffman E, Maeder ML, Khayter C, Reyon D, Dobbs D, Langenau DM, Stupar RM, Giraldez AJ, Voytas DF, Peterson RT, Yeh JRJ, Joung JK, Selection-free zinc-finger-nuclease engineering by context-dependent assembly (CoDA), Nat. Methods 8 (2011) 67–69. 10.1038/NMETH.1542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P, Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription, Nat. Biotechnol 29 (2011) 149–154. 10.1038/NBT.1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE, Church GM, RNA-guided human genome engineering via Cas9, Science. 339 (2013) 823–826. 10.1126/SCIENCE.1232033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Suwa T, Nyska A, Haseman JK, Mahler JF, Maronpot RR, Spontaneous lesions in control B6C3F1 mice and recommended sectioning of male accessory sex organs, Toxicol. Pathol 30 (2002) 228–234. 10.1080/019262302753559560. [DOI] [PubMed] [Google Scholar]
- [24].Grabowska MM, Degraff DJ, Yu X, Jin RJ, Chen Z, Borowsky AD, Matusik RJ, Mouse models of prostate cancer: picking the best model for the question, Cancer Metastasis Rev. 33 (2014) 377–397. 10.1007/S10555-013-9487-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Shappell SB, Thomas GV, Roberts RL, Herbert R, Ittmann MM, Rubin MA, Humphrey PA, Sundberg JP, Rozengurt N, Barrios R, Ward JM, Cardiff RD, 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, Cancer Res. 64 (2004) 2270–2305. 10.1158/0008-5472.CAN-03-0946. [DOI] [PubMed] [Google Scholar]
- [26].Oliveira DSM, Dzinic S, Bonfil AI, Saliganan AD, Sheng S, Bonfil RD, The mouse prostate: a basic anatomical and histological guideline., Bosn. J. Basic Med. Sci 16 (2016) 8–13. 10.17305/BJBMS.2016.917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Selman SH, The McNeal prostate: a review, Urology. 78 (2011) 1224–1228. 10.1016/J.UROLOGY.2011.07.1395. [DOI] [PubMed] [Google Scholar]
- [28].Toivanen R, Shen MM, Prostate organogenesis: tissue induction, hormonal regulation and cell type specification, Development. 144 (2017) 1382–1398. 10.1242/DEV.148270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Maser RS, Choudhury B, Campbell PJ, Feng B, Wong KK, Protopopov A, O’Neil J, Gutierrez A, Ivanova E, Perna I, Lin E, Mani V, Jiang S, McNamara K, Zaghlul S, Edkins S, Stevens C, Brennan C, Martin ES, Wiedemeyer R, Kabbarah O, Nogueira C, Histen G, Aster J, Mansour M, Duke V, Foroni L, Fielding AK, Goldstone AH, Rowe JM, Wang YA, Look AT, Stratton MR, Chin L, Futreal PA, DePinho RA, Chromosomally unstable mouse tumours have genomic alterations similar to diverse human cancers, Nature. 447 (2007) 966. 10.1038/NATURE05886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Rosenthal N, Brown S, The mouse ascending: perspectives for human-disease models, Nat. Cell Biol 9 (2007) 993–999. 10.1038/NCB437. [DOI] [PubMed] [Google Scholar]
- [31].Crowley L, Cambuli F, Aparicio L, Shibata M, Robinson BD, Xuan S, Li W, Hibshoosh H, Loda M, Rabadan R, Shen MM, A single-cell atlas of the mouse and human prostate reveals heterogeneity and conservation of epithelial progenitors, Elife. 9 (2020) 1–24. 10.7554/ELIFE.59465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Ittmann M, Anatomy and Histology of the Human and Murine Prostate, Cold Spring Harb. Perspect. Med 8 (2018). 10.1101/CSHPERSPECT.A030346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Inoue K, Fry EA, Taneja P, Recent progress in mouse models for tumor suppressor genes and its implications in human cancer, Clin. Med. Insights Oncol 7 (2013) 103–122. 10.4137/CMO.S10358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Maroulakou IG, Anver M, Garrett L, Green JE, Prostate and mammary adenocarcinoma in transgenic mice carrying a rat C3(1) simian virus 40 large tumor antigen fusion gene, Proc. Natl. Acad. Sci. U. S. A 91 (1994) 11236–11240. 10.1073/PNAS.91.23.11236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Zhang J, Thomas TZ, Kasper S, Matusik RJ, A small composite probasin promoter confers high levels of prostate-specific gene expression through regulation by androgens and glucocorticoids in vitro and in vivo, Endocrinology. 141 (2000) 4698–4710. 10.1210/endo.141.12.7837. [DOI] [PubMed] [Google Scholar]
- [36].Cleutjens KBJM, Van Der Korput HAGM, Ehren-van Eekelen CC, Sikes RA, Fasciana C, Chung LW, Trapman J, A 6-kb promoter fragment mimics in transgenic mice the prostate-specific and androgen-regulated expression of the endogenous prostate-specific antigen gene in humans, Mol. Endocrinol 11 (1997) 1256–1265. 10.1210/MEND.11.9.9974. [DOI] [PubMed] [Google Scholar]
- [37].Lee SH, Jia S, Zhu Y, Utermark T, Signoretti S, Loda M, Schaffhausen B, Roberts TM, Transgenic Expression of Polyomavirus Middle T Antigen in the Mouse Prostate Gives Rise to Carcinoma, J. Virol 85 (2011) 5581. 10.1128/JVI.02609-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Kasper S, Rennie PS, Bruchovsky N, Sheppard PC, Cheng H, Lin L, Shiu RPC, Snoek R, Matusik RJ, Cooperative binding of androgen receptors to two DNA sequences is required for androgen induction of the probasin gene, J. Biol. Chem 269 (1994) 31763–31769. 10.1016/s0021-9258(18)31761-7. [DOI] [PubMed] [Google Scholar]
- [39].Rennie PS, Bruchovsky N, Leco KJ, Sheppard PC, McQueen SA, Cheng H, Snoek R, Hamel A, Bock ME, MacDonald BS, Nickel BE, Chang C, Liao S, Cattini PA, Matusik RJ, Characterization of two cis-acting DNA elements involved in the androgen regulation of the probasin gene, Mol. Endocrinol 7 (1993) 23–36. 10.1210/MEND.7.1.8446105. [DOI] [PubMed] [Google Scholar]
- [40].Greenberg NM, DeMayo FJ, Sheppard PC, Barrios R, Lebovitz R, Finegold M, Angelopoulou R, Dodd JG, Duckworth ML, Rosen JM, Matusik RJ, The rat probasin gene promoter directs hormonally and developmentally regulated expression of a heterologous gene specifically to the prostate in transgenic mice, Mol. Endocrinol 8 (1994) 230–239. 10.1210/me.8.2.230. [DOI] [PubMed] [Google Scholar]
- [41].Yan Y, Sheppard PC, Kasper S, Lin L, Hoare S, Kapoor A, Dodd JG, Duckworth ML, Matusik RJ, Large Fragment of the Probasin Promoter Targets High Levels of Transgene Expression to the Prostate of Transgenic Mice, (1997). 10.1002/(SICI)1097-0045(19970701)32:2. [DOI] [PubMed] [Google Scholar]
- [42].Rotondo JC, Mazzoni E, Bononi I, Tognon M, Martini F, Association Between Simian Virus 40 and Human Tumors, Front. Oncol 9 (2019) 670. 10.3389/FONC.2019.00670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Ahuja D, Sáenz-Robles MT, Pipas JM, SV40 large T antigen targets multiple cellular pathways to elicit cellular transformation, Oncogene. 24 (2005) 7729–7745. 10.1038/SJ.ONC.1209046. [DOI] [PubMed] [Google Scholar]
- [44].Ratineau C, Ronco A, Leiter AB, Role of the amino-terminal domain of simian virus 40 early region in inducing tumors in secretin-expressing cells in transgenic mice., Gastroenterology. 119 (2000) 1305–1311. 10.1053/GAST.2000.19278. [DOI] [PubMed] [Google Scholar]
- [45].Pipas JM, Levine AJ, Role of T antigen interactions with p53 in tumorigenesis, Semin. Cancer Biol 11 (2001) 23–30. 10.1006/SCBI.2000.0343. [DOI] [PubMed] [Google Scholar]
- [46].Hernandez I, Maddison LA, Wei Y, Demayo F, Petras T, Li B, Gingrich JR, Rosen JM, Greenberg NM, Prostate-Specific Expression of p53 R172L Differentially Regulates p21, Bax, and mdm2 to Inhibit Prostate Cancer Progression and Prolong Survival, (2003). http://aacrjournals.org/mcr/article-pdf/1/14/1036/3131587/1036-1047.pdf (accessed April 17, 2024). [PubMed] [Google Scholar]
- [47].Sáenz-Robles MT, Sullivan CS, Pipas JM, Transforming functions of Simian Virus 40, Oncogene. 20 (2001) 7899–7907. 10.1038/sj.onc.1204936. [DOI] [PubMed] [Google Scholar]
- [48].Sontag E, Fedorov S, Kamibayashi C, Robbins D, Cobb M, Mumby M, The interaction of SV40 small tumor antigen with protein phosphatase 2A stimulates the map kinase pathway and induces cell proliferation, Cell. 75 (1993) 887–897. 10.1016/0092-8674(93)90533-V. [DOI] [PubMed] [Google Scholar]
- [49].Shibata MA, Jorcyk CL, Liu ML, Yoshidome K, Gold LG, Green JE, The C3(1)/SV40 T antigen transgenic mouse model of prostate and mammary cancer, Toxicol. Pathol 26 (1998) 177–182. 10.1177/019262339802600121. [DOI] [PubMed] [Google Scholar]
- [50].Garabedian EM, Humphrey PA, Gordon JI, A transgenic mouse model of metastatic prostate cancer originating from neuroendocrine cells, Proc. Natl. Acad. Sci 95 (1998) 15382–15387. 10.1073/PNAS.95.26.15382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Calvo A, Perez-Stable C, Segura V, Catena R, Guruceaga E, Nguewa P, Blanco D, Parada L, Reiner T, Green JE, Molecular characterization of the Gγ-globin-tagtransgenic mouse model of hormone refractory prostate cancer: comparison to human prostate cancer, Prostate. 70 (2010) 630–645. 10.1002/pros.21097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Perez-Stable C, Altman NH, Brown J, Harbison M, Cray C, Roos BA, Prostate, adrenocortical, and brown adipose tumors in fetal globin/T antigen transgenic mice., Lab. Invest 74 (1996) 363–373. https://europepmc.org/article/med/8780156 (accessed January 29, 2023). [PubMed] [Google Scholar]
- [53].Skalnik DG, Dorfman DM, Williams DA, Orkin SH, Restriction of neuroblastoma to the prostate gland in transgenic mice., Mol. Cell. Biol 11 (1991) 4518. 10.1128/MCB.11.9.4518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Kitsberg DI, Leder P, Keratinocyte growth factor induces mammary and prostatic hyperplasia and mammary adenocarcinoma in transgenic mice., Oncogene. 13 (1996) 2507–2515. https://europepmc.org/article/med/9000125 (accessed January 29, 2023). [PubMed] [Google Scholar]
- [55].Gingrich JR, Barrios RJ, Foster BA, Greenberg NM, Pathologic progression of autochthonous prostate cancer in the TRAMP model, Prostate Cancer Prostatic Dis. 2 (1999) 70–75. 10.1038/SJ.PCAN.4500296. [DOI] [PubMed] [Google Scholar]
- [56].Gingrich JR, Greenberg NM, A Transgenic Mouse Prostate Cancer Model, Toxicol. Pathol 24 (1996) 502–504. 10.1177/019262339602400414/ASSET/019262339602400414.FP.PNG_V03. [DOI] [PubMed] [Google Scholar]
- [57].Greenberg NM, Demayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, Cunha GR, Donjacour AA, Matusik RJ, Rosen JM, Prostate cancer in a transgenic mouse, Proc. Natl. Acad. Sci 92 (1995) 3439–3443. 10.1073/PNAS.92.8.3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Sharma P, Schreiber-Agus N, Mouse models of prostate cancer, Oncogene. 18 (1999) 5349–5355. 10.1038/sj.onc.1203037. [DOI] [PubMed] [Google Scholar]
- [59].Kaplan-Lefko PJ, Chen TM, Ittmann MM, Barrios RJ, Ayala GE, Huss WJ, Maddison LA, Foster BA, Greenberg NM, Pathobiology of autochthonous prostate cancer in a pre-clinical transgenic mouse model, Prostate. 55 (2003) 219–237. 10.1002/PROS.10215. [DOI] [PubMed] [Google Scholar]
- [60].Gingrich J, Barrios R, Kattan M, Nahm H, Finegold M, Greenberg N, Androgen-independent prostate cancer progression in the TRAMP model., Cancer Res. (1997). [PubMed] [Google Scholar]
- [61].Gingrich JR, Barrios RJ, Morton RA, Boyce BF, DeMayo FJ, Finegold MJ, Angelopoulou R, Rosen JM, Greenberg NM, Metastatic prostate cancer in a transgenic mouse, Cancer Res. 56 (1996) 4096–4102. https://pubmed.ncbi.nlm.nih.gov/8797572/ (accessed January 28, 2023). [PubMed] [Google Scholar]
- [62].Gelman IH, How the TRAMP Model Revolutionized the Study of Prostate Cancer Progression, Cancer Res. 76 (2016) 6137–6139. 10.1158/0008-5472.CAN-16-2636. [DOI] [PubMed] [Google Scholar]
- [63].Patel SJ, Molinolo AA, Gutkind S, Crawford NPS, Germline genetic variation modulates tumor progression and metastasis in a mouse model of neuroendocrine prostate carcinoma., PLoS One. 8 (2013) e61848. 10.1371/JOURNAL.PONE.0061848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Shui X, Xu R, Zhang C, Meng H, Zhao J, Shi C, Advances in neuroendocrine prostate cancer research: From model construction to molecular network analyses, Lab. Investig 2021 1024. 102 (2021) 332–340. 10.1038/s41374-021-00716-0. [DOI] [PubMed] [Google Scholar]
- [65].Tang Y, Wang L, Goloubeva O, Khan MA, Lee DI, Hussain A, The relationship of neuroendocrine carcinomas to anti-tumor therapies in TRAMP mice, Prostate. 69 (2009) 1763–1773. 10.1002/PROS.21026. [DOI] [PubMed] [Google Scholar]
- [66].Niu Y, Altuwaijri S, Yeh S, Lai KP, Yu S, Chuang KH, Huang SP, Lardy H, Chang C, Targeting the stromal androgen receptor in primary prostate tumors at earlier stages, Proc. Natl. Acad. Sci. U. S. A 105 (2008) 12188. 10.1073/PNAS.0804701105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Chiaverotti T, Couto SS, Donjacour A, Mao JH, Nagase H, Cardiff RD, Cunha GR, Balmain A, Dissociation of epithelial and neuroendocrine carcinoma lineages in the transgenic adenocarcinoma of mouse prostate model of prostate cancer, Am. J. Pathol 172 (2008) 236–246. 10.2353/AJPATH.2008.070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Gao F, Alwhaibi A, Sabbineni H, Verma A, Eldahshan W, Somanath PR, Suppression of Akt1-β-catenin pathway in advanced prostate cancer promotes TGFβ1-mediated epithelial to mesenchymal transition and metastasis, Cancer Lett. 402 (2017) 177–189. 10.1016/J.CANLET.2017.05.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Tepaamorndech S, Huang L, Kirschke CP, A null-mutation in the Znt7 gene accelerates prostate tumor formation in a transgenic adenocarcinoma mouse prostate model, Cancer Lett. 308 (2011) 33–42. 10.1016/J.CANLET.2011.04.011. [DOI] [PubMed] [Google Scholar]
- [70].Kasper S, Sheppard P, Yan Y, Pettigrew N, Borowsky A, Prins G, Dodd J, Duckworth ML, Matusik R, Development, progression, and androgen-dependence of prostate tumors in probasin-large T antigen transgenic mice: a model for prostate cancer., Lab. Invest (1998). [Google Scholar]
- [71].Masumori N, Thomas TZ, Chaurand P, Case T, Paul M, Kasper S, Caprioli RM, Tsukamoto T, Shappell SB, Matusik RJ, A Probasin-Large T Antigen Transgenic Mouse Line Develops Prostate Adenocarcinoma and Neuroendocrine Carcinoma with Metastatic Potential, Cancer Res. 61 (2001) 2239–2249. http://aacrjournals.org/cancerres/article-pdf/61/5/2239/2493254/ch050102239.pdf (accessed January 28, 2023). [PubMed] [Google Scholar]
- [72].Mordan-McCombs S, Brown T, Wang WLW, Gaupel AC, Welsh JE, Tenniswood M, Tumor Progression in the LPB-Tag Transgenic Model of Prostate Cancer is Altered by Vitamin D Receptor and Serum Testosterone Status, J. Steroid Biochem. Mol. Biol 121 (2010) 368. 10.1016/J.JSBMB.2010.03.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Zhou Z, Flesken-Nikitin A, Corney DC, Wang W, Goodrich DW, Roy-Burman P, Nikitin AY, Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer, Cancer Res. 66 (2006) 7889–7898. 10.1158/0008-5472.CAN-06-0486. [DOI] [PubMed] [Google Scholar]
- [74].Shui X, Xu R, Zhang C, Meng H, Zhao J, Shi C, Advances in neuroendocrine prostate cancer research: From model construction to molecular network analyses, Lab. Investig 2021 1024. 102 (2021) 332–340. 10.1038/s41374-021-00716-0. [DOI] [PubMed] [Google Scholar]
- [75].Kido LA, de Almeida Lamas C, Maróstica MR, Cagnon VHA, Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model: A good alternative to study PCa progression and chemoprevention approaches, Life Sci. 217 (2019) 141–147. 10.1016/J.LFS.2018.12.002. [DOI] [PubMed] [Google Scholar]
- [76].Irshad S, Abate-Shen C, Modeling prostate cancer in mice: Something old, something new, something premalignant, something metastatic, Cancer Metastasis Rev. 32 (2013) 109. 10.1007/S10555-012-9409-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Sternberg N, Demonstration and analysis of P1 site-specific recombination using lambda-P1 hybrid phages constructed in vitro, Cold Spring Harb. Symp. Quant. Biol 43 Pt 2 (1979) 1143–1146. 10.1101/SQB.1979.043.01.128. [DOI] [PubMed] [Google Scholar]
- [78].Metzger D, Chambon P, Site- and time-specific gene targeting in the mouse, Methods. 24 (2001) 71–80. 10.1006/METH.2001.1159. [DOI] [PubMed] [Google Scholar]
- [79].Abdulkadir SA, Magee JA, Peters TJ, Kaleem Z, Naughton CK, Humphrey PA, Milbrandt J, Conditional loss of Nkx3.1 in adult mice induces prostatic intraepithelial neoplasia, Mol. Cell. Biol 22 (2002) 1495–1503. 10.1128/MCB.22.5.1495-1503.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Ma X, Ziel-Van Der Made AC, Autar B, Van Der Korput HA, Vermeij M, Van Duijn P, Cleutjens KB, De Krijger R, Krimpenfort P, Berns A, Van Der Kwast TH, 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, Cancer Res. 65 (2005) 5730–5739. 10.1158/0008-5472.CAN-04-4519. [DOI] [PubMed] [Google Scholar]
- [81].Korsten H, Ziel-Van der Made ACJ, van Weerden WM, van der Kwast T, Trapman J, van Duijn PW, Characterization of Heterogeneous Prostate Tumors in Targeted Pten Knockout Mice, PLoS One. 11 (2016) e0147500. 10.1371/JOURNAL.PONE.0147500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Bierie B, Nozawa M, Renou JP, Shillingford JM, Morgan F, Oka T, Taketo MM, Cardiff RD, Miyoshi K, Wagner KU, Robinson GW, Hennighausen L, Activation of beta-catenin in prostate epithelium induces hyperplasias and squamous transdifferentiation, Oncogene. 22 (2003) 3875–3887. 10.1038/SJ.ONC.1206426. [DOI] [PubMed] [Google Scholar]
- [83].Backman SA, Ghazarian D, So K, Sanchez O, Wagner KU, Hennighausen L, Suzuki A, Tsao MS, Chapman WB, Stambolic V, Mak TW, Early onset of neoplasia in the prostate and skin of mice with tissue-specific deletion of Pten, Proc. Natl. Acad. Sci 101 (2004) 1725–1730. 10.1073/PNAS.0308217100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, Washington MK, Neilson EG, Moses HL, TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia, Science. 303 (2004) 848–851. 10.1126/SCIENCE.1090922. [DOI] [PubMed] [Google Scholar]
- [85].Wang X, De Julio MK, Economides KD, Walker D, Yu H, Halili MV, Hu YP, Price SM, Abate-Shen C, Shen MM, A luminal epithelial stem cell that is a cell of origin for prostate cancer, Nature. 461 (2009) 495–500. 10.1038/NATURE08361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Chow LML, Baker SJ, PTEN function in normal and neoplastic growth, Cancer Lett. 241 (2006) 184–196. 10.1016/J.CANLET.2005.11.042. [DOI] [PubMed] [Google Scholar]
- [87].Dahia PLM, PTEN, a unique tumor suppressor gene, Endocr. Relat. Cancer 7 (2000) 115–129. 10.1677/ERC.0.0070115. [DOI] [PubMed] [Google Scholar]
- [88].Podsypanina K, Ellenson LH, Nemes A, Gu J, Tamura M, Yamada KM, Cordon-Cardo C, Catoretti G, Fisher PE, Parsons R, Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems, Proc. Natl. Acad. Sci 96 (1999) 1563–1568. 10.1073/PNAS.96.4.1563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP, Pten is essential for embryonic development and tumour suppression, Nat. Genet 19 (1998) 348–355. 10.1038/1235. [DOI] [PubMed] [Google Scholar]
- [90].Trotman LC, Niki M, Dotan ZA, Koutcher JA, Di Cristofano A, Xiao A, Khoo AS, Roy-Burman P, Greenberg NM, Van Dyke T, Cordon-Cardo C, Pandolfi PP, Pten dose dictates cancer progression in the prostate, PLoS Biol. 1 (2003). 10.1371/JOURNAL.PBIO.0000059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Kwabi-Addo B, Giri D, Schmidt K, Podsypanina K, Parsons R, Greenberg N, Ittmann M, Haploinsufficiency of the Pten tumor suppressor gene promotes prostate cancer progression, Proc. Natl. Acad. Sci 98 (2001) 11563–11568. 10.1073/PNAS.201167798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Zhao G, Forn-Cuní G, Scheers M, Lindenbergh PP, Yin J, van Loosen Q, Passarini L, Chen L, Snaar-Jagalska BE, Simultaneous targeting of AMPK and mTOR is a novel therapeutic strategy against prostate cancer, Cancer Lett. (2024) 216657. 10.1016/J.CANLET.2024.216657. [DOI] [PubMed] [Google Scholar]
- [93].Bertram J, Peacock JW, Fazli L, Mui ALF, Chung SW, Cox ME, Monia B, Gleave ME, Ong CJ, Loss of PTEN is associated with progression to androgen independence, Prostate. 66 (2006) 895–902. 10.1002/PROS.20411. [DOI] [PubMed] [Google Scholar]
- [94].Liu S, Zhang B, Rowan BG, Jazwinski SM, Abdel-Mageed AB, Steele C, Wang AR, Sartor O, Niu T, Zhang Q, A Novel Controlled PTEN-Knockout Mouse Model for Prostate Cancer Study, Front. Mol. Biosci 8 (2021) 474. 10.3389/FMOLB.2021.696537/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, Thomas GV, Li G, Roy-Burman P, Nelson PS, Liu X, Wu H, Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer, Cancer Cell. 4 (2003) 209–221. 10.1016/S1535-6108(03)00215-0. [DOI] [PubMed] [Google Scholar]
- [96].Ittmann M, Huang J, Radaelli E, Martin P, Signoretti S, Sullivan R, Simons BW, Ward JM, Robinson BD, Chu GC, Loda M, Thomas G, Borowsky A, Cardiff RD, Animal models of human prostate cancer: the consensus report of the New York meeting of the Mouse Models of Human Cancers Consortium Prostate Pathology Committee, Cancer Res. 73 (2013) 2718–2736. 10.1158/0008-5472.CAN-12-4213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Gao H, Ouyang X, Banach-Petrosky W, Borowsky AD, Lin Y, Kim M, Lee H, Shih WJ, Cardiff RD, Shen MM, Abate-Shen C, A critical role for p27kip1 gene dosage in a mouse model of prostate carcinogenesis, Proc. Natl. Acad. Sci 101 (2004) 17204–17209. 10.1073/PNAS.0407693101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Hubbard GK, Mutton LN, Khalili M, McMullin RP, Hicks JL, Bianchi-Frias D, Horn LA, Kulac I, Moubarek MS, Nelson PS, Yegnasubramanian S, De Marzo AM, Bieberich CJ, Combined MYC Activation and Pten Loss Are Sufficient to Create Genomic Instability and Lethal Metastatic Prostate Cancer, Cancer Res. 76 (2016) 283–292. 10.1158/0008-5472.CAN-14-3280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Aytes A, Mitrofanova A, Kinkade CW, Lefebvre C, Lei M, Phelan V, LeKaye HC, Koutcher JA, Cardiff RD, Califano A, Shen MM, Abate-Shen C, ETV4 promotes metastasis in response to activation of PI3-kinase and Ras signaling in a mouse model of advanced prostate cancer, Proc. Natl. Acad. Sci 110 (2013). 10.1073/PNAS.1303558110/-/DCSUPPLEMENTAL/SD05.XLSX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Ding Z, Wu CJ, Chu GC, Xiao Y, Ho D, Zhang J, Perry SR, Labrot ES, Wu X, Lis R, Hoshida Y, Hiller D, Hu B, Jiang S, Zheng H, Stegh AH, Scott KL, Signoretti S, Bardeesy N, Wang YA, Hill DE, Golub TR, Stampfer MJ, Wong WH, Loda M, Mucci L, Chin L, Depinho RA, SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression, Nature. 470 (2011) 269–276. 10.1038/NATURE09677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Lee SH, Poulogiannis G, Pyne S, Jia S, Zou L, Signoretti S, Loda M, Cantley LC, Roberts TM, A constitutively activated form of the p110beta isoform of PI3-kinase induces prostatic intraepithelial neoplasia in mice, Proc. Natl. Acad. Sci 107 (2010) 11002–11007. 10.1073/PNAS.1005642107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Arriaga JM, Panja S, Alshalalfa M, Zhao J, Zou M, Giacobbe A, Madubata CJ, Kim JY, Rodriguez A, Coleman I, Virk RK, Hibshoosh H, Ertunc O, Ozbek B, Fountain J, Jeffrey Karnes R, Luo J, Antonarakis ES, Nelson PS, Feng FY, Rubin MA, De Marzo AM, Rabadan R, Sims PA, Mitrofanova A, Abate-Shen C, A MYC and RAS co-activation signature in localized prostate cancer drives bone metastasis and castration resistance, Nat. Cancer 1 (2020) 1082–1096. 10.1038/S43018-020-00125-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Zhang W, Dong Y, Sartor O, Zhang K, Deciphering the Increased Prevalence of TP53 Mutations in Metastatic Prostate Cancer, Cancer Inform. 21 (2022). 10.1177/11769351221087046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Elgavish A, Wood PA, Pinkert CA, Eltoum IE, Cartee T, Wilbanks J, Mentor-Marcel R, Tian L, Scroggins SE, Transgenic mouse with human mutant p53 expression in the prostate epithelium, Prostate. 61 (2004) 26–34. 10.1002/PROS.20071. [DOI] [PubMed] [Google Scholar]
- [105].Cho H, Herzka T, Zheng W, Qi J, Wilkinson JE, Bradner JE, Robinson BD, Castillo-Martin M, Cordon-Cardo C, Trotman LC, RapidCaP, a novel GEM model for metastatic prostate cancer analysis and therapy, reveals myc as a driver of Pten-mutant metastasis, Cancer Discov. 4 (2014) 319–333. 10.1158/2159-8290.CD-13-0346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Yong C, Moose DL, Bannick N, Gutierrez WR, Vanneste M, Svensson R, Breheny P, Brown JA, Dodd RD, Cohen MB, Henry MD, Locally invasive, castrate-resistant prostate cancer in a Pten/Trp53 double knockout mouse model of prostate cancer monitored with non-invasive bioluminescent imaging, PLoS One. 15 (2020). 10.1371/JOURNAL.PONE.0232807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Martin P, Liu YN, Pierce R, Abou-Kheir W, Casey O, Seng V, Camacho D, Simpson RM, Kelly K, Prostate Epithelial Pten/TP53 Loss Leads to Transformation of Multipotential Progenitors and Epithelial to Mesenchymal Transition, Am. J. Pathol 179 (2011) 422. 10.1016/J.AJPATH.2011.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Vinall RL, Chen JQ, Hubbard NE, Sulaimon SS, Shen MM, DeVere White RW, Borowsky AD, Initiation of prostate cancer in mice by Tp53R270H: evidence for an alternative molecular progression, Dis. Model. Mech 5 (2012) 914–920. 10.1242/DMM.008995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Zou M, Toivanen R, Mitrofanova A, Floch N, Hayati S, Sun Y, Le Magnen C, Chester D, Mostaghel EA, Califano A, Rubin MA, Shen MM, Abate-Shen C, Transdifferentiation as a Mechanism of Treatment Resistance in a Mouse Model of Castration-Resistant Prostate Cancer, Cancer Discov. 7 (2017) 736–749. 10.1158/2159-8290.CD-16-1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Ding Z, Wu CJ, Jaskelioff M, Ivanova E, Kost-Alimova M, Protopopov A, Chu GC, Wang G, Lu X, Labrot ES, Hu J, Wang W, Xiao Y, Zhang H, Zhang J, Zhang J, Gan B, Perry SR, Jiang S, Li L, Horner JW, Wang YA, Chin L, Depinho RA, Telomerase reactivation following telomere dysfunction yields murine prostate tumors with bone metastases, Cell. 148 (2012) 896–907. 10.1016/j.cell.2012.01.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].He WW, Sciavolino PJ, Wing J, Augustus M, Hudson P, Meissner PS, Curtis RT, Shell BK, Bostwick DG, Tindall DJ, Gelmann EP, Abate-Shen C, Carter KC, A Novel Human Prostate-Specific, Androgen-Regulated Homeobox Gene (NKX3.1) That Maps to 8p21, a Region Frequently Deleted in Prostate Cancer, Genomics. 43 (1997) 69–77. 10.1006/GENO.1997.4715. [DOI] [PubMed] [Google Scholar]
- [112].Abate-Shen C, Shen MM, Gelmann E, Integrating differentiation and cancer: The Nkx3.1 homeobox gene in prostate organogenesis and carcinogenesis, Differentiation. 76 (2008) 717. 10.1111/J.1432-0436.2008.00292.X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Bhatia-Gaur R, Donjacour AA, Sciavolino PJ, Kim M, Desai N, Young P, Norton CR, Gridley T, Cardiff RD, Cunha GR, Abate-Shen C, Shen MM, Roles for Nkx3.1 in prostate development and cancer, Genes Dev. 13 (1999) 966–977. 10.1101/GAD.13.8.966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Schneider A, Brand T, Zweigerdt R, Arnold HH, Targeted disruption of the Nkx3.1 gene in mice results in morphogenetic defects of minor salivary glands: Parallels to glandular duct morphogenesis in prostate, Mech. Dev 95 (2000) 163–174. 10.1016/S0925-4773(00)00355-5. [DOI] [PubMed] [Google Scholar]
- [115].Bowen C, Bubendorf L, Voeller HJ, Slack R, Willi N, Sauter G, Gasser TC, Koivisto P, Lack EE, Kononen J, Kallioniemi OP, Gelmann EP, Loss of NKX3.1 expression in human prostate cancers correlates with tumor progression, Cancer Res. 60 (2000) 6111–6115. [PubMed] [Google Scholar]
- [116].Kim M, Bhatia-Gaur R, Banach-Petrosky WA, Desai N, Wang Y, Hayward S, Cunha G, Cardiff R, Shen M, Abate-Shen C, Nkx3.1 mutant mice recapitulate early stages of prostate carcinogenesis., Cancer Res. (2002). [PubMed] [Google Scholar]
- [117].Kim MJ, Cardiff RD, Desai N, Banach-Petrosky WA, Parsons R, Shen MM, Abate-Shen C, Cooperativity of Nkx3.1 and Pten loss of function in a mouse model of prostate carcinogenesis, Proc. Natl. Acad. Sci 99 (2002) 2884–2889. 10.1073/PNAS.042688999/ASSET/234C5B7D-5DD8-4736-BCA3-81671E999D30/ASSETS/GRAPHIC/PQ0426889005.JPEG. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Bowen C, Ostrowski MC, Leone G, Gelmann EP, Loss of PTEN Accelerates NKX3.1 Degradation to Promote Prostate Cancer Progression, Cancer Res. 79 (2019) 4124–4134. 10.1158/0008-5472.CAN-18-4110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Yao Y, Gu X, Xu X, Ge S, Jia R, Novel insights into RB1 mutation, Cancer Lett. 547 (2022). 10.1016/J.CANLET.2022.215870. [DOI] [PubMed] [Google Scholar]
- [120].Hill R, Song Y, Cardiff RD, Van Dyke T, Heterogeneous Tumor Evolution Initiated by Loss of pRb Function in a Preclinical Prostate Cancer Model, Cancer Res. 65 (2005) 10243–10254. 10.1158/0008-5472.CAN-05-1579. [DOI] [PubMed] [Google Scholar]
- [121].Maddison LA, Sutherland BW, Barrios RJ, Greenberg NM, Conditional deletion of Rb causes early stage prostate cancer, Cancer Res. 64 (2004) 6018–6025. 10.1158/0008-5472.CAN-03-2509. [DOI] [PubMed] [Google Scholar]
- [122].Ku SY, Rosario S, Wang Y, Mu P, Seshadri M, Goodrich ZW, Goodrich MM, Labbé DP, Gomez EC, Wang J, Long HW, Xu B, Brown M, Loda M, Sawyers CL, Ellis L, Goodrich DW, Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance, Science. 355 (2017). 10.1126/SCIENCE.AAH4199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Ellwood-Yen K, Graeber TG, Wongvipat J, Iruela-Arispe ML, Zhang JF, Matusik R, Thomas GV, Sawyers CL, Myc-driven murine prostate cancer shares molecular features with human prostate tumors, Cancer Cell. 4 (2003) 223–238. 10.1016/S1535-6108(03)00197-1. [DOI] [PubMed] [Google Scholar]
- [124].Iwata T, Schultz D, Hicks J, Hubbard GK, Mutton LN, Lotan TL, Bethel C, Lotz MT, Yegnasubramanian S, Nelson WG, Dang CV, Xu MM, Anele U, Koh CM, Bieberich CJ, De Marzo AM, MYC Overexpression Induces Prostatic Intraepithelial Neoplasia and Loss of Nkx3.1 in Mouse Luminal Epithelial Cells, PLoS One. 5 (2010) e9427. 10.1371/JOURNAL.PONE.0009427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Lee JK, Phillips JW, Smith BA, Park JW, Stoyanova T, McCaffrey EF, Baertsch R, Sokolov A, Meyerowitz JG, Mathis C, Cheng D, Stuart JM, Shokat KM, Gustafson WC, Huang J, Witte ON, N-Myc Drives Neuroendocrine Prostate Cancer Initiated from Human Prostate Epithelial Cells, Cancer Cell. 29 (2016) 536–547. 10.1016/J.CCELL.2016.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Dardenne E, Beltran H, Benelli M, Gayvert K, Berger A, Puca L, Cyrta J, Sboner A, Noorzad Z, MacDonald T, Cheung C, Yuen KS, Gao D, Chen Y, Eilers M, Mosquera JM, Robinson BD, Elemento O, Rubin MA, Demichelis F, Rickman DS, N-Myc Induces an EZH2-Mediated Transcriptional Program Driving Neuroendocrine Prostate Cancer, Cancer Cell. 30 (2016) 563–577. 10.1016/j.ccell.2016.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Stiles B, Gilman V, Khanzenzon N, Lesche R, Li A, Qiao R, Liu X, Wu H, Essential role of AKT-1/protein kinase B alpha in PTEN-controlled tumorigenesis, Mol. Cell. Biol 22 (2002) 3842–3851. 10.1128/MCB.22.11.3842-3851.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Majumder PK, Yeh JJ, George DJ, Febbo PG, Kum J, Xue Q, Bikoff R, Ma H, Kantoff PW, Golub TR, Loda M, Sellers WR, Prostate intraepithelial neoplasia induced by prostate restricted Akt activation: The MPAKT model, Proc. Natl. Acad. Sci. U. S. A 100 (2003) 7841. 10.1073/PNAS.1232229100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Qian C, Li D, Chen Y, ETS factors in prostate cancer, Cancer Lett. 530 (2022) 181–189. 10.1016/J.CANLET.2022.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Tomlins SA, Laxman B, Varambally S, Cao X, Yu J, Helgeson BE, Cao Q, Prensner JR, Rubin MA, Shah RB, Mehra R, Chinnaiyan AM, Role of the TMPRSS2-ERG Gene Fusion in Prostate Cancer, Neoplasia. 10 (2008) 177. 10.1593/NEO.07822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Demichelis F, Fall K, Perner S, Andrén O, Schmidt F, Setlur SR, Hoshida Y, Mosquera JM, Pawitan Y, Lee C, Adami HO, Mucci LA, Kantoff PW, Andersson SO, Chinnaiyan AM, Johansson JE, Rubin MA, TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort, Oncogene. 26 (2007) 4596–4599. 10.1038/SJ.ONC.1210237. [DOI] [PubMed] [Google Scholar]
- [132].Delliaux C, Tian TV, Bouchet M, Fradet A, Vanpouille N, Flourens A, Deplus R, Villers A, Leroy X, Clézardin P, de Launoit Y, Bonnelye E, Duterque-Coquillaud M, TMPRSS2:ERG gene fusion expression regulates bone markers and enhances the osteoblastic phenotype of prostate cancer bone metastases, Cancer Lett. 438 (2018) 32–43. 10.1016/J.CANLET.2018.08.027. [DOI] [PubMed] [Google Scholar]
- [133].Klezovitch O, Risk M, Coleman I, Lucas JM, Null M, True LD, Nelson PS, Vasioukhin V, A causal role for ERG in neoplastic transformation of prostate epithelium, Proc. Natl. Acad. Sci 105 (2008) 2105–2110. https://doi.org/10.1073兾pnas.0711711105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Tomlins SA, Laxman B, Dhanasekaran SM, Helgeson BE, Cao X, Morris DS, Menon A, Jing X, Cao Q, Han B, Yu J, Wang L, Montie JE, Rubin MA, Pienta KJ, Roulston D, Shah RB, Varambally S, Mehra R, Chinnaiyan AM, Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer, Nature. 448 (2007) 595–599. 10.1038/NATURE06024. [DOI] [PubMed] [Google Scholar]
- [135].Carver BS, Tran J, Gopalan A, Chen Z, Shaikh S, Carracedo A, Alimonti A, Nardella C, Varmeh S, Scardino PT, Cordon-Cardo C, Gerald W, Pandolfi PP, Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate, Nat. Genet 41 (2009) 619–624. 10.1038/NG.370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].King JC, Xu J, Wongvipat J, Hieronymus H, Carver BS, Leung DH, Taylor BS, Sander C, Cardiff RD, Couto SS, Gerald WL, Sawyers CL, Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis, Nat. Genet 41 (2009) 524–526. 10.1038/NG.371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Boysen G, Barbieri CE, Prandi D, Blattner M, Chae SS, Dahija A, Nataraj S, Huang D, Marotz C, Xu L, Huang J, Lecca P, Chhangawala S, Liu D, Zhou P, Sboner A, Debono JS, Demichelis F, Houvras Y, Rubin MA, SPOP mutation leads to genomic instability in prostate cancer, Elife. 4 (2015). 10.7554/ELIFE.09207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Blattner M, Liu D, Robinson BD, Huang D, Poliakov A, Gao D, Nataraj S, Deonarine LD, Augello MA, Sailer V, Ponnala L, Ittmann M, Chinnaiyan AM, Sboner A, Chen Y, Rubin MA, Barbieri CE, SPOP Mutation Drives Prostate Tumorigenesis In Vivo through Coordinate Regulation of PI3K/mTOR and AR Signaling, Cancer Cell. 31 (2017) 436–451. 10.1016/J.CCELL.2017.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Shenoy TR, Boysen G, Wang MY, Xu QZ, Guo W, Koh FM, Wang C, Zhang LZ, Wang Y, Gil V, Aziz S, Christova R, Rodrigues DN, Crespo M, Rescigno P, Tunariu N, Riisnaes R, Zafeiriou Z, Flohr P, Yuan W, Knight E, Swain A, Ramalho-Santos M, Xu DY, de Bono J, Wu H, CHD1 loss sensitizes prostate cancer to DNA damaging therapy by promoting error-prone double-strand break repair, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol 28 (2017) 1495–1507. 10.1093/ANNONC/MDX165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Rescigno P, Boysen G, Rodrigues DN, Seed G, Dolling D, Riisnaes R, Crespo M, Bianchini D, Sumanasuriya S, Figueiredo I, Christova R, Gil V, Goodall J, Sharp A, Rubin MA, Yuan W, Barbieri C, Mateo J, Carreira S, De Bono JS, Molecular and clinical implications of CHD1 loss and SPOP mutations in advanced prostate cancer. 36 (2018) 5064–5064. 10.1200/JCO.2018.36.15_SUPPL.5064. [DOI] [Google Scholar]
- [141].Hernández-Llodrà S, Segalés L, Safont A, Juanpere N, Lorenzo M, Fumadó L, Rodríguez-Vida A, Cecchini L, Bellmunt J, Lloreta-Trull J, SPOP and FOXA1 mutations are associated with PSA recurrence in ERG wt tumors, and SPOP downregulation with ERG-rearranged prostate cancer, Prostate. 79 (2019) 1156–1165. 10.1002/PROS.23830. [DOI] [PubMed] [Google Scholar]
- [142].Adams EJ, Karthaus WR, Hoover E, Liu D, Gruet A, Zhang Z, Cho H, DiLoreto R, Chhangawala S, Liu Y, Watson PA, Davicioni E, Sboner A, Barbieri CE, Bose R, Leslie CS, Sawyers CL, FOXA1 mutations alter pioneering activity, differentiation, and prostate cancer phenotypes, Nature. 571 (2019) 408. 10.1038/S41586-019-1318-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Malaney P, Nicosia SV, Davé V, One mouse, one patient paradigm: New avatars of personalized cancer therapy, Cancer Lett. 344 (2014) 1–12. 10.1016/J.CANLET.2013.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Kersten K, de Visser KE, van Miltenburg MH, Jonkers J, Genetically engineered mouse models in oncology research and cancer medicine, EMBO Mol. Med 9 (2017) 137. 10.15252/EMMM.201606857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Schmidt DR, Monique I, Gramatikov T, Sheen A, Williams CL, Hurwitz M, Dodge LE, Holupka E, Kiger Iii WS, Cornwall-Brady MR, Huang W, Mak HH, Cormier KS, Condon C, Wittrup KD, Yilmaz ÖH, Stevenson MA, Down JD, Floyd SR, Roper J, Vander Heiden MG, Ablative radiotherapy improves survival but does not cure autochthonous mouse models of prostate and colorectal cancer, Commun. Med 2023 31. 3 (2023) 1–20. 10.1038/s43856-023-00336-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Wang C, Zhang Y, Gao WQ, The evolving role of immune cells in prostate cancer, Cancer Lett. 525 (2022) 9–21. 10.1016/J.CANLET.2021.10.027. [DOI] [PubMed] [Google Scholar]
- [147].Kang J, La Manna F, Bonollo F, Sampson N, Alberts IL, Mingels C, Afshar-Oromieh A, Thalmann GN, Karkampouna S, Tumor microenvironment mechanisms and bone metastatic disease progression of prostate cancer, Cancer Lett. 530 (2022) 156–169. 10.1016/J.CANLET.2022.01.015. [DOI] [PubMed] [Google Scholar]
- [148].Flores-Téllez T. del N.J., Baena E, Experimental challenges to modeling prostate cancer heterogeneity, Cancer Lett. 524 (2022) 194–205. 10.1016/J.CANLET.2021.10.012. [DOI] [PubMed] [Google Scholar]
- [149].Jamroze A, Chatta G, Tang DG, Androgen receptor (AR) heterogeneity in prostate cancer and therapy resistance, Cancer Lett. 518 (2021) 1–9. 10.1016/J.CANLET.2021.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Mai CW, Chin KY, Foong LC, Pang KL, Yu B, Shu Y, Chen S, Cheong SK, Chua CW, Modeling prostate cancer: What does it take to build an ideal tumor model?, Cancer Lett. 543 (2022) 215794. 10.1016/J.CANLET.2022.215794. [DOI] [PubMed] [Google Scholar]
- [151].Monaco G, Van Dam S, Casal Novo Ribeiro JL, Larbi A, De Magalhães JP, A comparison of human and mouse gene co-expression networks reveals conservation and divergence at the tissue, pathway and disease levels, BMC Evol. Biol 15 (2015). 10.1186/S12862-015-0534-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Shoag J, Barbieri C, Clinical variability and molecular heterogeneity in prostate cancer, Asian J. Androl 18 (2016) 543. 10.4103/1008-682X.178852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Zhang D, Zhao S, Li X, Kirk JS, Tang DG, Prostate Luminal Progenitor Cells in Development and Cancer, Trends in Cancer. 4 (2018) 769. 10.1016/J.TRECAN.2018.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Crowley L, Shen MM, Heterogeneity and complexity of the prostate epithelium: New findings from single-cell RNA sequencing studies, Cancer Lett. 525 (2022) 108–114. 10.1016/J.CANLET.2021.10.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Yoshidome K, Shibata MA, Maroulakou IG, Liu ML, Jorcyk CL, Gold L, Welch VN, Green JE, Genetic alterations in the development of mammary and prostate cancer in the C3(1)/Tag transgenic mouse model (review), Int. J. Oncol 12 (1998) 449–453. 10.3892/IJO.12.2.449/HTML. [DOI] [PubMed] [Google Scholar]
- [156].Shibata M, Ward J, Devor D, Liu ML, Green J, Progression of prostatic intraepithelial neoplasia to invasive carcinoma in C3(1)/SV40 large T antigen transgenic mice: histopathological and molecular biological alterations., Cancer Res. (1996). [PubMed] [Google Scholar]
- [157].Tehranian A, Morris DW, Min H, Bird DJ, Cardiff RD, Barry PA, Neoplastic Transformation of Prostatic And Urogenital Epithelium by the Polyoma Virus Middle T Gene, Am. J. Pathol 149 (1996) 1177–1191. [PMC free article] [PubMed] [Google Scholar]
- [158].Zhang X, Chen MW, Ng A, Ng PY, Lee C, Rubin M, Olsson CA, Buttyan R, Abnormal prostate development in C3(1)-bcl-2 transgenic mice., Prostate. 32 (1997) 16–26. . [DOI] [PubMed] [Google Scholar]
- [159].Donjacour AA, Thomson AA, Cunha GR, Enlargement of the ampullary gland and seminal vesicle, but not the prostate in int-2/Fgf-3 transgenic mice, Differentiation. 62 (1998) 227–237. 10.1046/J.1432-0436.1998.6250227.X. [DOI] [PubMed] [Google Scholar]
- [160].Hennighausen L, McKnight R, Burdon T, Baik M, Wall RJ, Smith GH, Whey acidic protein extrinsically expressed from the mouse mammary tumor virus long terminal repeat results in hyperplasia of the coagulation gland epithelium and impaired mammary development., Cell Growth Differ. 5 (1994) 607–613. https://europepmc.org/article/med/7522033 (accessed January 29, 2023). [PubMed] [Google Scholar]
- [161].Maddison LA, Nahm H, Demayo F, Greenberg NM, Prostate specific expression of Cre recombinase in transgenic mice, Genesis. 26 (2000) 154–156. . [DOI] [PubMed] [Google Scholar]
- [162].Wang S, Garcia AJ, Wu M, Lawson DA, Witte ON, Wu H, Pten deletion leads to the expansion of a prostatic stem/progenitor cell subpopulation and tumor initiation, Proc. Natl. Acad. Sci. U. S. A 103 (2006) 1480–1485. 10.1073/pnas.0510652103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Jin C, McKeehan K, Wang F, Transgenic mouse with high Cre recombinase activity in all prostate lobes, seminal vesicle, and ductus deferens, Prostate. 57 (2003) 160–164. 10.1002/pros.10283. [DOI] [PubMed] [Google Scholar]
- [164].Ratnacaram CK, 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, Proc. Natl. Acad. Sci. U. S. A 105 (2008) 2521–2526. 10.1073/pnas.0712021105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Luchman HA, Friedman HC, Villemaire ML, Peterson AC, Jirik FR, Temporally controlled prostate epithelium-specific gene alterations, Genesis. 46 (2008) 229–234. 10.1002/dvg.20386. [DOI] [PubMed] [Google Scholar]
- [166].Birbach A, Casanova E, Schmid JA, A Probasin-MerCreMer BAC allows inducible recombination in the mouse prostate, Genesis. 47 (2009) 757–764. 10.1002/DVG.20558. [DOI] [PubMed] [Google Scholar]
- [167].Di Cristofano A, De Acetis M, Koff A, Cordon-Cardo C, P Pandolfi P, Pten and p27KIP1 cooperate in prostate cancer tumor suppression in the mouse, Nat. Genet 27 (2001) 222–224. 10.1038/84879. [DOI] [PubMed] [Google Scholar]
- [168].Nandana S, Ellwood-Yen K, Sawyers C, Wills M, Weidow B, Case T, Vasioukhin V, Matusik R, Hepsin cooperates with MYC in the progression of adenocarcinoma in a prostate cancer mouse model, Prostate. 70 (2010) 591–600. 10.1002/PROS.21093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Jin RJ, Lho Y, Connelly L, Wang Y, Yu X, Saint Jean L, Case TC, Ellwood-Yen K, Sawyers CL, Bhowmick NA, Blackwell TS, Yull FE, Matusik RJ, The nuclear factor-κB pathway controls the progression of prostate cancer to androgen-independent growth, Cancer Res. 68 (2008) 6762–6769. 10.1158/0008-5472.CAN-08-0107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Kim J, Eltoum IEA, Roh M, Wang J, Abdulkadir SA, Interactions between cells with distinct mutations in c-MYC and Pten in prostate cancer, PLoS Genet. 5 (2009). 10.1371/JOURNAL.PGEN.1000542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Anderson PD, McKissic SA, Logan M, Roh M, Franco OE, Wang J, Doubinskaia I, Van Der Meer R, Hayward SW, Eischen CM, Eltoum IE, Abdulkadir SA, Nkx3.1 and Myc crossregulate shared target genes in mouse and human prostate tumorigenesis, J. Clin. Invest 122 (2012) 1907–1919. 10.1172/JCI58540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Kim J, Roh M, Doubinskaia I, Algarroba GN, Eltoum IEA, Abdulkadir SA, A mouse model of heterogeneous, c-MYC-initiated prostate cancer with loss of Pten and p53, Oncogene. 31 (2012) 322–332. 10.1038/ONC.2011.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].DeGraff DJ, Grabowska MM, Case TC, Yu X, Herrick MK, Hayward WJ, Strand DW, Cates JM, Hayward SW, Gao N, Walter MA, Buttyan R, Yi Y, Kaestner KH, Matusik RJ, FOXA1 deletion in luminal epithelium causes prostatic hyperplasia and alteration of differentiated phenotype, Lab. Investig 2014 947. 94 (2014) 726–739. 10.1038/labinvest.2014.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Stanbrough M, Leav I, Kwan PWL, Bubley GJ, Balk SP, Prostatic intraepithelial neoplasia in mice expressing an androgen receptor transgene in prostate epithelium, Proc. Natl. Acad. Sci 98 (2001) 10823–10828. 10.1073/PNAS.191235898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Han G, Buchanan G, Ittmann M, Harris JM, Yu X, DeMayo FJ, Tilley W, Greenberg NM, Mutation of the androgen receptor causes oncogenic transformation of the prostate, Proc. Natl. Acad. Sci. U. S. A 102 (2005) 1151. 10.1073/PNAS.0408925102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Liu G, Sprenger C, Sun S, Epilepsia KS, Haugk K, Zhang X, Coleman I, Nelson PS, Plymate S, AR Variant ARv567es Induces Carcinogenesis in a Novel Transgenic Mouse Model of Prostate Cancer, Neoplasia. 15 (2013) 1009–IN3. 10.1593/NEO.13784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Zhu C, Luong R, Zhuo M, Johnson DT, McKenney JK, Cunha GR, Sun Z, Conditional expression of the androgen receptor induces oncogenic transformation of the mouse prostate, J. Biol. Chem 286 (2011) 33478–33488. 10.1074/JBC.M111.269894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Bruxvoort KJ, Charbonneau HM, Giambernardi TA, Goolsby JC, Qian CN, Zylstra CR, Robinson DR, Roy-Burman P, Shaw AK, Buckner-Berghuis BD, Sigler RE, Resau JH, Sullivan R, Bushman W, Williams BO, Inactivation of Apc in the mouse prostate causes prostate carcinoma, Cancer Res. 67 (2007) 2490–2496. 10.1158/0008-5472.CAN-06-3028. [DOI] [PubMed] [Google Scholar]
- [179].Valkenburg KC, Yu X, De Marzo AM, Spiering TJ, Matusik RJ, Williams BO, Activation of Wnt/β-catenin signaling in a subpopulation of murine prostate luminal epithelial cells induces high grade prostate intraepithelial neoplasia, Prostate. 74 (2014) 1506–1520. 10.1002/pros.22868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Yu X, Wang Y, Jiang M, Bierie B, Roy-Burman P, Shen MM, Taketo MM, Wills M, Matusik RJ, Activation of β-Catenin in mouse prostate causes HGPIN and continuous prostate growth after castration, Prostate. 69 (2009) 249–262. 10.1002/pros.20877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Francis JC, Thomsen MK, Taketo MM, Swain A, β-Catenin Is Required for Prostate Development and Cooperates with Pten Loss to Drive Invasive Carcinoma, PLoS Genet. 9 (2013). 10.1371/journal.pgen.1003180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Pearson HB, Phesse TJ, Clarke AR, K-ras and Wnt signaling synergize to accelerate prostate tumorigenesis in the mouse, Cancer Res. 69 (2009) 94–101. 10.1158/0008-5472.CAN-08-2895. [DOI] [PubMed] [Google Scholar]
- [183].Nopparat J, Zhang J, Lu JP, Chen YH, Zheng D, Neufer PD, Fan JM, Hong H, Boykin C, Lu Q, δ-Catenin, a Wnt/β-catenin modulator, reveals inducible mutagenesis promoting cancer cell survival adaptation and metabolic reprogramming, Oncogene. 34 (2014) 1542–1552. 10.1038/onc.2014.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Yu X, Wang Y, Degraff DJ, Wills ML, Matusik RJ, Wnt/β-Catenin activation promotes prostate tumor progression in a mouse model, Oncogene. 30 (2011) 1868. 10.1038/ONC.2010.560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Qin J, Wu SP, Creighton CJ, Dai F, Xie X, Cheng CM, Frolov A, Ayala G, Lin X, Feng XH, Ittmann MM, Tsai SJ, Tsai MJ, Tsai SY, COUP-TFII inhibits TGF-β-induced growth barrier to promote prostate tumorigenesis, Nat. 2012 4937431. 493 (2012) 236–240. 10.1038/nature11674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Mulholland DJ, Kobayashi N, Ruscetti M, Zhi A, Tran LM, Huang J, Gleave M, Wu H, Pten loss and RAS/MAPK activation cooperate to promote EMT and metastasis initiated from prostate cancer stem/progenitor cells, Cancer Res. 72 (2012) 1878–1889. 10.1158/0008-5472.CAN-11-3132/650317/AM/PTEN-LOSS-AND-RAS-MAPK-ACTIVATION-COOPERATE-TO. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Jeong JH, Wang Z, Guimaraes AS, Ouyang X, Figueiredo JL, Ding Z, Jiang S, Guney I, Kang GH, Shin E, Hahn WC, Loda MF, Abate-Shen C, Weissleder R, Chin L, BRAF Activation Initiates but Does Not Maintain Invasive Prostate Adenocarcinoma, PLoS One. 3 (2008) e3949. 10.1371/JOURNAL.PONE.0003949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Qin J, Lee HJ, Wu SP, Lin SC, Lanz RB, Creighton CJ, DeMayo FJ, Tsai SY, Tsai MJ, Androgen deprivation–induced NCoA2 promotes metastatic and castration-resistant prostate cancer, J. Clin. Invest 124 (2014) 5013–5026. 10.1172/JCI76412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Li N, Xue W, Yuan H, Dong B, Ding Y, Liu Y, Jiang M, Kan S, Sun T, Ren J, Pan Q, Li X, Zhang P, Hu G, Wang Y, Wang X, Li Q, Qin J, AKT-mediated stabilization of histone methyltransferase WHSC1 promotes prostate cancer metastasis, J. Clin. Invest 127 (2017) 1284–1302. 10.1172/JCI91144. [DOI] [PMC free article] [PubMed] [Google Scholar]