Figure 3 - uploaded by Anil Kapoor
Content may be subject to copyright.
Prostate cancer initiation, progression and advancement are associated with proto-oncogene activation and inhibition of tumor suppressor genes involved in PI3K/AKT, RAS/MAPK and STAT3 signaling. Abnormalities in the prostate epithelium result in pre-neoplastic lesions called prostatic intraepithelial neoplasia (PIN), which feature luminal epithelial hyperplasia and a reduction in the number of basal cells. PIN lesions progress to invasive adenocarcinoma (luminal phenotype) with loss of the basal cell layer and basement membrane resulting in various tumor grades, beginning with indolent to more aggressive forms of PCa, and subsequent development of metastasis and castration resistance. Studies involving murine PCa models provide support for the role of proteins involved in PI3K/AKT, RAS/MAPK and STAT3 signaling at various stages of PCa development. Murine PCa models discussed in the text are illustrated, with specific promoter-driven gene knockout or transgene overexpression indicated in brackets. Figure is adapted from ref. [5]. BM bone marrow; c-Akt constitutive-active Akt; CR castration-resistant; Cre Cre-recombinase; CreER/CreERT2 TAM-inducible Cre; LN lymph node; OT orthotopic; PB probasin promoter; TAM tamoxifen. 

Prostate cancer initiation, progression and advancement are associated with proto-oncogene activation and inhibition of tumor suppressor genes involved in PI3K/AKT, RAS/MAPK and STAT3 signaling. Abnormalities in the prostate epithelium result in pre-neoplastic lesions called prostatic intraepithelial neoplasia (PIN), which feature luminal epithelial hyperplasia and a reduction in the number of basal cells. PIN lesions progress to invasive adenocarcinoma (luminal phenotype) with loss of the basal cell layer and basement membrane resulting in various tumor grades, beginning with indolent to more aggressive forms of PCa, and subsequent development of metastasis and castration resistance. Studies involving murine PCa models provide support for the role of proteins involved in PI3K/AKT, RAS/MAPK and STAT3 signaling at various stages of PCa development. Murine PCa models discussed in the text are illustrated, with specific promoter-driven gene knockout or transgene overexpression indicated in brackets. Figure is adapted from ref. [5]. BM bone marrow; c-Akt constitutive-active Akt; CR castration-resistant; Cre Cre-recombinase; CreER/CreERT2 TAM-inducible Cre; LN lymph node; OT orthotopic; PB probasin promoter; TAM tamoxifen. 

Source publication
Article
Full-text available
The cells of the prostate gland are dependent on cell signaling pathways to regulate their growth, maintenance and function. However, perturbations in key signaling pathways, resulting in neoplastic transformation of cells in the prostate epithelium, are likely to generate subtypes of prostate cancer which may subsequently require different treatme...

Context in source publication

Context 1
... to propagate PCSCs derived from human PCa cell lines including suspension or low adherence culture [67, 68], and media cultures containing either fetal bovine serum [60, 61, 66] or serum-free media supplemented with epidermal growth factor [75, 76] and basic fibroblast growth factor [67, 69, 70, 72]. However, the formation of free-floating spheres under serum-free conditions is routinely used as an in vitro approach towards propagating and evaluating the stem-like properties of putative CSCs derived from solid tumors [62], including those derived from PCa cells [69, 70, 72]. Despite its limitations [77], the sphere formation assay allows for the clonality and self-renewal capacity of stem-like cells to be evaluated upon culturing individual cells, reducing the impact of progenitor-like cells which display limited self-renewal potential [78]. The self-renewal capacity of sphere-derived human PCSCs has been evaluated [70, 76], which display enhanced tumor formation in vivo [69, 70, 72, 76]. While purification of human PCa cells based on the expression of surface markers isolates stem-like cells with tumor-initiating ability (Table 1), flow cytometry can also enrich for prospective PCSCs based on their ability to efflux the Hoechst 33342 dye. These cells, also known as side population (SP) cells [79], isolated from androgen-sensitive LAPC9 cells are capable of in vivo tumor formation unlike the non-SP cell population [80]. The SP phenotype is associated with the expression of the ATP-binding cassette membrane transporter family of proteins, particularly ABCG2 [81], which are involved in drug efflux and confer multi-drug resistance [82]. However, ABCG2 + and ABCG2 - PCa cells displayed similar tumorigenic potential in vivo [80], implicating the expression of additional ABC transporter proteins and/or other means of Hoechst 33342 dye efflux by these PCa cells. An alternative strategy towards purifying PCSCs involves measuring their aldehyde dehydrogenase (ALDH) activity. Elevated ALDH activity has been associated with CSCs in various solid tumors [83] and is required to maintain the drug-tolerant cancer cell subpopulation [84]. ALDH1A1, which is expressed by a minor basal cell population in human prostates [85], was observed to be heterogeneously expressed in prostate adenocarcinomas, with elevated ALDH1A1 expression being positively correlated with the severity (Gleason score and pathologic stage) of the disease and inversely correlated with patient survival [85, 86]. Primary and metastatic PCa also express ALDH7A1 protein [87], with knockdown of ALDH7A1 reducing the tumor-propagating and metastatic abilities of androgen-independent PCa cells [88]. Using a functional (AldefluorTM) assay validated to assess ALDH1 isoform activities [89, 90], PCa cells demonstrating high ALDH activity (ALDH hi ) were isolated and displayed increased tumorigenicity in vivo [87]. In addition, as few as 10 1 ALDH hi CD44 + α 2 β 1+ cells isolated from LAPC9 (low PSA- expressing; PSA low ) tumor xenografts generated secondary tumors in castrated mice whereas 10 4 ALDH low CD44 - α 2 β 1- cells were required for tumor formation, indicating that the ALDH hi CD44 + α 2 β 1+ phenotype in a PSA low cell population is enriched for castration-resistant PCSCs [91]. While 19 ALDH isoforms have been identified in humans [83], other ALDH isoforms have yet to be shown to be functionally- active in PCSCs or required for tumor formation in vivo . As no individual method isolates bona-fide human PCSCs exclusively, a combination of methods (sphere culture, and isolation based on cell surface marker expression and/or functional activity) can be used to enrich for and characterize human PCSCs, with subsequent implantation in immunocompromised mice to evaluate their tumor- propagating activity and serial transplantation to assess their long-term self-renewal capability in vivo [62]. A number of cell signaling pathways have been implicated in PCa progression towards an androgen- resistant state including receptor tyrosine kinases, like epidermal growth factor receptor (EGFR) [92-94], and developmental pathways including Wnt, Notch and Hedgehog signaling [95-97]. These developmental pathways have also been implicated in PCSC maintenance [98-100]. Human PSCs and PCSCs display low levels or lack AR expression [45, 64, 66, 72], with evidence to suggest that murine PSCs and PCSCs are capable of surviving in an androgen-independent state [26, 32, 101]. While not to undermine the importance of these developmental pathways and AR signaling in PCa, we will focus on the involvement of intracellular PI3K/AKT, RAS/MAPK and STAT3 pathways in maintaining PCSCs. Given that details of these signaling pathways and their ability to regulate various hallmarks of PCa have been reviewed thoroughly elsewhere [102-104], we highlight key proteins within these pathways that regulate PCSC activity (Figure 2), and the animal models that support their role in PCa development (Figure 3). Dysregulation of PI3K/AKT signaling has been implicated in PCa given the discovery that its negative regulator, PTEN, is mutated and frequently lost in PCa. Loss of PTEN function, as a result of mutation, deletion or reduced expression, occurs at a frequency of ~40% in PCa [105, 106]. Integrative analysis of mRNA expression, copy number and exon sequencing for somatic mutations conducted on a cohort of PCa patients revealed that the PI3K/AKT pathway was altered in 42% of primary cancers and 100% of metastases, while the RAS/MAPK pathway was altered in 43% of primary specimens and 90% of metastases [106]. Both PTEN loss and AKT activation, as measured by Ser 473 phosphorylation, are associated with biochemical relapse following radical prostatectomy [107, 108], while PTEN deletion (along with allelic c-MYC gain) is associated with biochemical relapse following local therapy [59]. Intense staining patterns for activated AKT are correlated with higher Gleason grade PCa [109] and tumor progression [110], while PTEN loss is associated with PCa progression and predicts a shorter time for metastasis-free survival in patients [111]. The role of PI3K/AKT signaling in PCa has also been examined in animal models. Pten +/- mice were first reported to display hyperplastic lesions in the prostate, with benign PIN lesions being observed in some mice at a young age (≤14 weeks) [112, 113]. Pten deletion in prostate basal or luminal cells has also been carried out to promote PI3K/AKT signal activation in order to determine the prostate cell lineage responsible for subsequent tumor initiation [26, 32, 35], as previously discussed. Prior to these studies, probasin ( PB ) promoter-driven Pten deletion was conducted, resulting in PI3K/AKT signal activation specifically in both prostate basal and luminal cells [114]. By 6 weeks of age, these mice formed PIN lesions, developed adenocarcinoma that invades stromal regions (9 weeks) and displays androgen-independent survival following castration, and generated metastases in the lymph nodes and lungs (12 weeks) [114]. Moreover, prostate epithelial cells lacking Pten require mammalian target of rapamycin (mTOR) complex 2 (mTORC2) for neoplastic transformation [115], thereby implicating downstream mTOR signaling in murine PCa development. Expression of a constitutively-active form of Akt1 (c-Akt) resulted in PIN formation when expression is driven by the PB promoter [116], or after reconstituting c-Akt- expressing prostate basal cells with UGSM cells and grafting under the kidney capsule of mice [49]. Therefore, the murine prostate epithelium has been shown to be vulnerable to Pten loss, with the propensity to undergo AKT-driven tumorigenesis. The role of PI3K/AKT signaling in the proliferation and maintenance of PCSCs has been examined in human PCa. PTEN knockdown in DU145 PCa cells resulted in an increased ability to enrich for CD133/AC141 + CD44 + stem-like cells [69]. Treatment with PI3K inhibitor LY294002 reduced sphere formation [69] while the dual PI3K/mTOR inhibitor NVP-BEZ235 reduced the CD133/ AC141 + CD44 + cell population in vivo and subsequently delayed tumor formation [117]. However, DU145 spheres display low levels of AKT activation while PTEN knockdown marginally increased AKT activation and did not affect sphere maintenance, suggesting that PTEN function may not be critical for PCSC self-renewal in vitro [70]. While PTEN may not be required to negatively- regulate PI3K/AKT-dependent PCSC maintenance, whether PTEN loss/inactivity and subsequent PI3K/AKT signal activation promotes PCSC differentiation into non- tumorigenic bulk tumor cells has not been thoroughly evaluated, despite PTEN knockdown increasing tumor formation in vivo [69]. Mitogen-activated protein kinases (MAPKs) are cytoplasmic Ser/threonine kinases that transduce signals by activating extracellular signal-regulated kinases (ERKs) through the RAS-RAF-MEK-ERK (RAS/MAPK) signal transduction cascade [118]. Similar to PI3K/AKT signaling, MAPK signal activation correlates with disease progression, with elevated staining for phosphorylated (active) forms of ERK1 and ERK2 (ERK1/2) being detected with increasing tumor stage of human PCa specimens [119]. Increased ERK1/2 activation in patients receiving neoadjuvant hormone therapy, and in recurrent CRPC patients, has been observed [120]. Mutations of all three RAS isoforms ( KRAS , HRAS and NRAS ) have been detected in human PCa specimens (3 – 30%), particularly among Japanese men [121-124], while the activating mutant B-RAF(V600E) has been observed in ~10% of Korean PCa patients [122]. These correlative studies reinforce that activated RAS/MAPK signaling contributes towards prostate tumorigenesis. Further evidence in support of a relationship between RAS/MAPK signaling and PCa progression towards an androgen-independent state has been demonstrated in the preclinical setting. Ectopic expression of HRAS(T35S) in androgen-sensitive LNCaP cells ...

Similar publications

Article
Full-text available
The PI3K/AKT pathway is frequently altered in advanced human prostate cancer mainly through the loss of functional PTEN, and presents as potential target for personalized therapy. Our aim was to determine the therapeutic potential of the pan-AKT inhibitor, AZD5363, in PTEN-deficient prostate cancer. Here we used a genetically engineered mouse (GEM)...
Article
Full-text available
Currently, no therapeutic options exist for castration-resistant prostate cancer (CRPC) patients who have developed resistance to the second generation of anti-androgen receptor (AR) axis therapy. Here we report that co-deletion of Pten and p53 in murine prostate epithelium, often observed in human CRPC, leads to AR-independent CRPC and thus confer...
Article
Full-text available
Genomic instability is a hallmark of cancer often associated with poor patient outcome and resistance to targeted therapy. Assessment of genomic instability in bulk tumor or biopsy can be complicated due to sample availability, surrounding tissue contamination, or tumor heterogeneity. The Epic Sciences circulating tumor cell (CTC) platform utilizes...
Article
Full-text available
Despite recent advances in the treatment for advanced prostate cancer, outcomes remain poor. This lack of efficacy has prompted the development of alternative treatment strategies. In the present study we investigate the effects of the multikinase inhibitor sorafenib in a genetically engineered mouse model of prostate cancer and explore the rationa...
Article
Full-text available
Aberrant elevation of JARID1B and histone H3 lysine 4 trimethylation (H3K4me3) is frequently observed in many diseases including prostate cancer (PCa), yet the mechanisms on the regulation of JARID1B and H3K4me3 through epigenetic alterations still remain poorly understood. Here we report that Skp2 modulates JARID1B and H3K4me3 levels in vitro in c...

Citations

... In addition to their tumorigenicity and high metastatic rate, PC-SCs isolated from either AR + or AR − PCa cell lines were observed to undergo epithelial-tomesenchymal (EMT) phenotypic transition, by displaying mesenchymal surface markers instead of epithelial markers. Many other CSC signaling pathways, including Wnt/β-Catenin, Notch, TGF-β, and Hedgehog, have been studied, shown to play important roles in the maintenance of PCSCs, and may contribute to PCa progression and recurrence [252,262]. ...
... Notably, NEPCs share certain molecular characteristics with PCSCs, such as lack of androgen receptor (AR−), retinoblastoma gene (RB−), and prostate-specific antigen (PSA − ), while they differ in specific expression of neuronal differentiation markers, such as chromogranin A, neuron-specific enolase (NSE), synaptophysin, tubulin beta III (TUBB3), TTF-1, and CD56 (NCAM) by NEPCs [262,281]. When comparing the NE cells in PCa tissue and those found in benign tissue, it is visible that the NE cells in the PCa tissue are morphologically different and display both epithelial markers and NE markers [278]. ...
Article
Full-text available
Simple Summary In this review paper, we explore the role of chronic inflammation in the pathogenesis of prostatic diseases, such as chronic prostatitis, benign prostatic hyperplasia (BPH), and prostate cancer. Chronic inflammation is a significant risk factor for these diseases, but the molecular mechanisms behind it are not yet fully understood. Previous attempts to investigate, link, and target inflammatory signaling molecules in men with these conditions have been clinically ambiguous and inconclusive. As a result, we proposed a paradigm shift in which less-explored molecules in the inflammatory signaling cascade are investigated as possible therapeutic targets for prostate diseases. In addition, we comprehensively discuss how aberrant signaling of these molecules may cause prostate cancer stemness, neuroendocrine differentiation, castration resistance, metabolic reprogramming, and immunosuppression. In conclusion, we forecast that targeting the interleukin-1 receptor-associated kinases (IRAKs) signaling pathway may provide a far more effective therapeutic or prophylactic strategy for the management of chronic inflammation-driven prostatic diseases. Abstract Chronic inflammation is now recognized as one of the major risk factors and molecular hallmarks of chronic prostatitis, benign prostatic hyperplasia (BPH), and prostate tumorigenesis. However, the molecular mechanisms by which chronic inflammation signaling contributes to the pathogenesis of these prostate diseases are poorly understood. Previous efforts to therapeutically target the upstream (e.g., TLRs and IL1-Rs) and downstream (e.g., NF-κB subunits and cytokines) inflammatory signaling molecules in people with these conditions have been clinically ambiguous and unsatisfactory, hence fostering the recent paradigm shift towards unraveling and understanding the functional roles and clinical significance of the novel and relatively underexplored inflammatory molecules and pathways that could become potential therapeutic targets in managing prostatic diseases. In this review article, we exclusively discuss the causal and molecular drivers of prostatitis, BPH, and prostate tumorigenesis, as well as the potential impacts of microbiome dysbiosis and chronic inflammation in promoting prostate pathologies. We specifically focus on the importance of some of the underexplored druggable inflammatory molecules, by discussing how their aberrant signaling could promote prostate cancer (PCa) stemness, neuroendocrine differentiation, castration resistance, metabolic reprogramming, and immunosuppression. The potential contribution of the IL1R-TLR-IRAK-NF-κBs signaling molecules and NLR/inflammasomes in prostate pathologies, as well as the prospective benefits of selectively targeting the midstream molecules in the various inflammatory cascades, are also discussed. Though this review concentrates more on PCa, we envision that the information could be applied to other prostate diseases. In conclusion, we have underlined the molecular mechanisms and signaling pathways that may need to be targeted and/or further investigated to better understand the association between chronic inflammation and prostate diseases.
... The PI3K-AKT-mTOR signaling axis has been shown to play a crucial role in the development and maintenance of CRPC [62][63][64]. This pathway integrates growth signals with downstream processes that increase cell survival and proliferation [64]. ...
Article
Full-text available
Dogs are one of few species that naturally develop prostate cancer (PCa), which clinically resembles aggressive, advanced PCa in humans. Moreover, PCa-tumor samples from dogs are often androgen receptor (AR)-negative and may enrich our understanding of AR-indifferent PCa in humans, a highly lethal subset of PCa for which few treatment modalities are available This narrative review discusses the molecular similarities between dog PCa and specific human-PCa variants, underscoring the possibilities of using the dog as a novel pre-clinical animal model for human PCa, resulting in new therapies and diagnostics that may benefit both species.
... Survival of PCSCs could promote tumor initiation, migration, and relapse, and induce therapy resistance (10,11). In addition, PCSC plasticity is anticipated to play a pivotal role in bone metastases of prostate cancer (12). ...
Article
Full-text available
The development of new therapeutic strategies is on the increase for prostate cancer stem cells, owing to current standardized therapies for prostate cancer, including chemotherapy, androgen deprivation therapy (ADT), radiotherapy, and surgery, often failing because of tumor relapse ability. Ultimately, tumor relapse develops into advanced castration-resistant prostate cancer (CRPC), which becomes an irreversible and systemic disease. Hence, early identification of the intracellular components and molecular networks that promote prostate cancer is crucial for disease management and therapeutic intervention. One of the potential therapeutic methods for aggressive prostate cancer is to target prostate cancer stem cells (PCSCs), which appear to be a primary focal point of cancer metastasis and recurrence and are resistant to standardized therapies. PCSCs have also been documented to play a major role in regulating tumorigenesis, sphere formation, and the metastasis ability of prostate cancer with their stemness features. Therefore, the current review highlights the origin and identification of PCSCs and their role in anti-androgen resistance, as well as stemness-related signaling pathways. In addition, the review focuses on the current advanced therapeutic strategies for targeting PCSCs that are helping to prevent prostate cancer initiation and progression, such as microRNAs (miRNAs), nanotechnology, chemotherapy, immunotherapy, the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) gene-editing system, and photothermal ablation (PTA) therapy.
... PCSCs are a subset of cells within prostate tumors; they have a long-term self-renewal potential and cause clinical treatment failure, tumor relapse, and metastasis [43]. Since JICD of JAG1 is involved in malignant cell transformation [44], we investigated whether JICD promotes PC stem-like cell properties in PC cells, leading to advanced PCs. ...
... CD133 has been considered the best exemplified and used marker of cancer stem cells (CSCs), including PCSCs [45,46]. Meanwhile, CD44 and CD133 have been used to identify basal prostate stem cells (PSCs) [43] because basal cells in the prostate epithelial layer are CD44 + p63 + CK5 + CK14 + . Normal PSCs are well known as CD44 + CD133 + AR −/low PSA −/low [43], while PCSCs are CD133 + AR high [45]. ...
... Meanwhile, CD44 and CD133 have been used to identify basal prostate stem cells (PSCs) [43] because basal cells in the prostate epithelial layer are CD44 + p63 + CK5 + CK14 + . Normal PSCs are well known as CD44 + CD133 + AR −/low PSA −/low [43], while PCSCs are CD133 + AR high [45]. ...
Article
Full-text available
JAG1 expression is upregulated in high-grade metastatic prostate carcinomas and associated with poor disease-free survival of patients with prostate cancer. Intriguingly, all JAG1-positive prostate carcinomas express JICD although JICD function in prostate cancer (PC) cells is poorly understood. In this study, we found that JICD overexpression increased the expression levels of AR, especially AR-Vs, in PC cell lines and significantly enhanced androgen-independent and androgen-dependent function of ARs. Interestingly, JICD overexpression upregulated the expression of the PCSC marker CD133 in PC cells as the expression of self-renewal markers; namely, NANOG and OCT3/4 increased. In addition, JICD overexpression highly increased the expression of anti-apoptotic BCL-XL protein, while it little affected the expression of apoptotic BIM protein. In 3D cell culture assays, the spheres formed by JICD-overexpressing PC subline cells (C4-2 and CWR22Rv1) were larger than those formed by control (EV) subline cells with undifferentiated morphology. Although JICD overexpression caused quiescence in cell proliferation, it activated the expression of components in PCSC-related signaling pathways, increased PC cell mobility, and promoted in vivo xenograft mouse tumorigenesis. Therefore, JICD may play a crucial role in enhancing androgen independence and promoting stem-like properties in PC cells and should be considered a novel target for CRPC and PCSC diagnostic therapy.
... These factors ultimately activate the proliferation of cells, suppress the apoptotic genes [166], and upregulate anti-apoptotic genes such as Beta2-Macroglobulin, B-Cell CLL/Lymphoma-2 (BCL2), and BCLXL [167]. The induction of STAT3 by IL-6 [168] or stress factors [169] leads to progress in the self-renewal of prostate CSCs and tumor-propagation [170]. VEGF binds to VEGFR-2/JAK2/STAT3, leading to the activation of STAT3 and upregulation of Myc and SOX2. ...
Article
Full-text available
Over the past two decades, cancer stem cells (CSCs) have emerged as an immensely studied and experimental topic, however a wide range of questions concerning the topic still remain unanswered; in particular, the mechanisms underlying the regulation of tumor stem cells and their characteristics. Understanding the cancer stem-cell signaling pathways may pave the way towards a better comprehension of these mechanisms. Signaling pathways such as WNT, STAT, Hedgehog, NOTCH, PI3K/AKT/mTOR, TGF-β, and NF-κB are responsible not only for modulating various features of CSCs but also their microenvironments. Recently, the prominent roles of various non-coding RNAs such as small non-coding RNAs (sncRNAs) and long non-coding RNAs (lncRNAs) in developing and enhancing the tumor phenotypes have been unfolded. This review attempts to shed light on understanding the influence of long non- coding RNAs in the modulation of various CSC-signaling pathways and its impact on the CSCs and tumor properties; highlighting the protagonistic and antagonistic roles of lncRNAs.
... Considering the fact that activation of STAT3 significantly correlates with poor prognosis in gastric cancer [9], tumor invasion, lymph node metastasis and tumor grade in colorectal cancer [10] and a higher risk of recurrence [11][12] in breast cancer, it is plausible that STAT3 activation is involved in CSCs regulation as in gastric cancer [13]. As it has been previously shown that downregulation of STAT3 activity inhibits the proliferation, colony expansion and clonogenic growth of stem-like cells in colorectal cancer [14][15] and its activation by IL-6 [16] or stress factors like ROS [17] results in enhanced self-renewal and tumorigenicity capacity of CSCs in the prostate [18][19], and glioblastoma [20]. Moreover, we recently found that STAT3 up-regulated in gastric stem-like cells [21]. ...
Article
Full-text available
Signal transducer and activator of transcription 3 (STAT3) has been recognized with dual effects in provision of cancer; either tumor inductive or immune suppressive. Recent findings considering the role of STAT3 in stem cells and cancer stem cell regulation, but its role in gastric cancer stem cells (GCSCs) and modulating the Th17/Treg balance is unknown. In the present study, we aimed to evaluate the role of activated STAT3 in GCSCs and Th17/ Treg cell paradigm. In completion of our previous results, the findings here indicate that gastro-spheroids, as a model of GCSCs, represent higher level of STAT3 activity, up-regulation of TGF-b and VEGF with downregulation of IL-6. On the other hand, treatment of normal naïve T cells with conditioned medium derived from gastro-spheroids promotes T cell differentiation toward cells with a higher level of FOXP3, TGF-b, and IL-10 expression which is indicative of Treg cells. Suppression of STAT3 activation in cancer cells by using Stattic small molecule treatment, decreases stemness features (i.e. spheroid formation and integrity, stemness gene expression and in vivo tumorigenicity capacity) and downregulates TGF-b in the cancer cells. Furthermore, co-culture of conditioned medium of STAT3 inhibited cancer cells with normal PBMCs leads to reduction in the percentage of Treg accompanied with increase of Th17 cells with a decrease in the secretion of TGF-b and increase in IFN-γ in T cells under differentiation. Therefore, targeting the STAT3 pathway in cancer cells seems to control the tumor formation and also impact on immune cells shifting to antitumor Th17 population.
... Most mCRPC, including both CRPC-adenocarcinoma (CRPC-adeno) and CRPC-NE subtypes, remains lethal. Figure is modified from [85] and [86]. Bottom: The trajectory of PCa development and progression is accompanied by loss of differentiation with increasing malignancy and aggressiveness at both cellular and molecular levels as well as at the tumor level. ...
... Neuroendocrine cells express neuroendocrine markers such as synaptophysin and chromogranin A and do not express AR [86]. The stroma is populated by fibroblasts, immune cells, smooth muscle cells and varies subtypes of neural cells, such as TH + , NES + , and β-Tubulin III + nerve fibers and abundant GFAP + cells [83]. Figure is modified from [85] and [83]. ...
Article
Full-text available
Cancer progression is characterized and driven by gradual loss of a differentiated phenotype and gain of stem cell-like features. In prostate cancer (PCa), androgen receptor (AR) signaling is important for cancer growth, progression, and emergence of therapy resistance. Targeting the AR signaling axis has been, over the decades, the mainstay of PCa therapy. However, AR signaling at the transcription level is reduced in high-grade cancer relative to low-grade PCa and loss of AR expression promotes a stem cell-like phenotype, suggesting that emergence of resistance to AR-targeted therapy may be associated with loss of AR signaling and gain of stemness. In the present mini-review, we first discuss PCa from the perspective of an abnormal organ with increasingly deregulated differentiation, and discuss the role of AR signaling during PCa progression. We then focus on the relationship between prostate cancer stem cells (PCSCs) and AR signaling. We further elaborate on the current methods of using transcriptome-based stemness-enriched signature to evaluate the degree of oncogenic dedifferentiation (cancer stemness) in pan-cancer datasets, and present the clinical significance of scoring transcriptome-based stemness across the spectrum of PCa development. Our discussions highlight the importance to evaluate the dynamic changes in both stem cell-like features (stemness score) and AR signaling activity across the PCa spectrum.
... This atypical augmentation of the latter pathway distorts the protein-serine/threonine kinases promoting castration-resistant growth. PI3K-AKT-mTOR pathway interacts and cooperates with several key oncogenic signaling cascades such as MAPK and WNT signaling to facilitate PCa growth and drug resistance [116] . Furthermore, augmented levels of ERKs activation have been reported in recurrent mCRPC patients, and correlative studies have further linked several mutations down the RAS isoforms with the tumorigeneses of PCa [117] . ...
Article
Full-text available
Prostate cancer (PCa) is a leading cause of cancer-related morbidity and mortality in men globally. Despite improvements in the diagnosis and treatment of PCa, a significant proportion of patients with high-risk localized disease and all patients with advanced disease at diagnosis will experience progression to metastatic castration-resistant prostate cancer (mCRPC). Multiple drugs are now approved as the standard of care treatments for patients with mCRPC that have been shown to prolong survival. Although the majority of patients will respond initially, primary and secondary resistance to these therapies make mCRPC an incurable disease. Several molecular mechanisms underlie the development of mCRPC, with the androgen receptor (AR) axis being the main driver as well as the key drug target. Understanding resistance mechanisms is crucial for discovering novel therapeutic strategies to delay or reverse the progression of the disease. In this review, we address the diverse mechanisms of drug resistance in mCRPC. In addition, we shed light on emerging targeted therapies currently being tested in clinical trials with promising potential to overcome mCRPC-drug resistance.
... TCGA, oncomine, nephroseq, and GEO (gene expression omnibus) are the most widely used databases in biological sciences 11 . These databases mainly GEO store vast amount of datasets related with cancer, diabetes, and other biological problems 8,[12][13][14][15][16] . The identification of pathogenetically distinct tumour types poses a significant challenge in the treatment of complex diseases (especially cancer) [17][18][19] . ...
Article
Full-text available
Cancer is among the highly complex disease and renal cell carcinoma is the sixth-leading cause of cancer death. In order to understand complex diseases such as cancer, diabetes and kidney diseases, high-throughput data are generated at large scale and it has helped in the research and diagnostic advancement. However, to unravel the meaningful information from such large datasets for comprehensive and minute understanding of cell phenotypes and disease pathophysiology remains a trivial challenge and also the molecular events leading to disease onset and progression are not well understood. With this goal, we have collected gene expression datasets from publicly available dataset which are for two different stages (I and II) for renal cell carcinoma and furthermore, the TCGA and cBioPortal database have been utilized for clinical relevance understanding. In this work, we have applied computational approach to unravel the differentially expressed genes, their networks for the enriched pathways. Based on our results, we conclude that among the most dominantly altered pathways for renal cell carcinoma, are PI3K-Akt, Foxo, endocytosis, MAPK, Tight junction, cytokine-cytokine receptor interaction pathways and the major source of alteration for these pathways are MAP3K13, CHAF1A, FDX1, ARHGAP26, ITGBL1, C10orf118, MTO1, LAMP2, STAMBP, DLC1, NSMAF, YY1, TPGS2, SCARB2, PRSS23, SYNJ1, CNPPD1, PPP2R5E. In terms of clinical significance, there are large number of differentially expressed genes which appears to be playing critical roles in survival.
... 10 CSCs also express various cell surface markers such as CD133, CD44, and/or integrin α 2 β 1 . 11 Experimental evidence suggests that PCSCs harboring CD44 markers show more tumor invasive properties. 11,12 Again, the presence of the androgen receptor (AR) can be associated with PCa with respect to its aggressiveness and therefore is further discussed elaborately in this Review. ...
... 11 Experimental evidence suggests that PCSCs harboring CD44 markers show more tumor invasive properties. 11,12 Again, the presence of the androgen receptor (AR) can be associated with PCa with respect to its aggressiveness and therefore is further discussed elaborately in this Review. PCSCs also distinctively and differentially express noncoding RNAs like miRNA (micro-RNA) and long-noncoding RNAs (lncRNAs) which synchronize to enhance the stemness of PCSCs. ...