There is evidence that cancer stem-like cells (CSC) and neuroendocrine behavior play critical roles in the pathogenesis and clinical course of metastatic castration-resistant prostate cancer (m-CRPC). However, there is limited mechanistic understanding of how CSC and neuroendocrine phenotypes impact the development of m-CRPC. In this study, we explored the role of the intracellular chemokine CXCL12γ in CSC induction and neuroendocrine differentiation and its impact on m-CRPC. CXCL12γ expression was detected in small-cell carcinoma of metastatic tissues and circulating tumor cells from m-CRPC patients and in prostate cancer cells displaying an neuroendocrine phenotype. Mechanistic investigations demonstrated that overexpression of CXCL12γ induced CSC and neuroendocrine phenotypes in prostate cancer cells through CXCR4-mediated PKCα/NFκB signaling, which promoted prostate tumor outgrowth, metastasis, and chemoresistance in vivo. Together, our results establish a significant function for CXCL12γ in m-CRPC development and suggest it as a candidate therapeutic target to control aggressive disease.

Significance: Expression of CXCL12γ induces the expression of a cancer stem cell and neuroendocrine phenotypes, resulting in the development of aggressive m-CRPC. Cancer Res; 78(8); 2026–39. ©2018 AACR.

Bone is the most frequent site of prostate cancer metastasis, and skeletal lesions are a common and disabling consequence of progressive disease (1, 2). First-line therapy for skeletal metastasis includes androgen ablation and chemotherapy. Yet, in the vast majority of cases, metastatic castration-resistant prostate cancer (m-CRPC) emerges and poses significant challenges for treatment (1–7). Unfortunately, many of the molecular mechanisms that lead to the development of m-CRPC remain unclear.

Growing evidence suggests that cancer stem cells (CSC) play a crucial role in cancer chemoresistance and recurrence in the development of m-CRPC (8–11). For example, after second-generation antiandrogens and chemotherapy, neuroendocrine carcinoma, potentially derived from CSCs, can accelerate metastatic castration-resistant progression, resulting in an aggressive clinical course (12–15). Thus, understanding the mechanisms by which CSCs and neuroendocrine differentiation impact the development of m-CRPC may facilitate the identification of biomarkers and development of novel therapeutics.

Stromal-derived factor-1 (SDF-1 or CXCL12) and CXCR4 signaling is known to play a central role in promoting tumor progression in many cancers and prostate cancer bone metastasis (16–23). The CXCL12–CXCR4 axis also directly promotes tumor cell motility based on chemokine gradients of CXCL12 and CXCR4 expression on tumor cells (21, 23). Recently, we demonstrated that enhanced levels of CXCL12 by cancer-associated fibroblasts (CAF) promote an epithelial–mesenchymal transition (EMT), supporting prostate cancer bone metastasis (18, 20). Moreover, CXCL12 signaling promotes activation of protein kinase C (PKC)/NFκB signaling, which contributes to CSC activities (24). These data provide direct evidence that CXCL12 plays a central role in the development of prostate cancer progression. To date, most studies have focused on the secreted isoforms of CXCL12 (CXCL12α, CXCL12β) as they promote metastasis and the survival of tumor cells (16–23). The role of the alternatively transcribed CXCL12γ isoform, an intracellular chemokine, in tumor progression remains unknown (25–30).

Here, the role of CXCL12γ expression in induction of CSC and neuroendocrine phenotypes in the development of m-CRPC was explored. These investigations demonstrate a unique and significant role of CXCL12γ in the reprogramming of CSCs and neuroendocrine cells, leading to the development of m-CRPC and reveal a potential central molecular pathway in the progression of aggressive disease.

Animals

Five- to 7-week-old male and female SCID mice (CB.17. SCID; Charles River Laboratories) were used as transplant recipients. All animal procedures were performed in compliance with the institutional ethical requirements and approved by the University of Michigan Institutional Committee for the Use and Care of Animals.

Cell culture

Human prostate cancer cell lines (PC3, DU145, and LNCaP) were obtained from the ATCC. The metastatic subclone of LNCaP, C4-2B, was originally isolated from a lymph node of a prostate cancer patient with disseminated bony and lymph node involvement. GFP-expressing prostate cancer cell lines (PC3GFP and DU145GFP cells) were established by lentiviral transduction. A castration-resistant subline of the LNCaP cells, LNCaP95, was kindly provided by Dr. Jun Luo (Johns Hopkins University, Baltimore, MD). All prostate cancer cell lines were routinely grown in RPMI1640 (Life Technologies) supplemented with 10% FBS (GEMINI Bio-Products), 1% penicillin/streptomycin (Life Technologies), and maintained at 37°C, 5% CO2, and 100% humidity. In some cases, prostate cancer cells were cultured in phenol red-free RPMI1640/DMEM (HyClone) supplemented with 10% charcoal/dextran-treated FBS (HyClone). Normal human prostate epithelial RWPE-1 cells (ATCC, CRL-11609) were cultured in Keratinocyte-SFM (Life Technologies) with supplements (17005-042, Life Technologies). Neuroendocrine prostate cancer cells (NCI-H660; ATCC, CRL 5813) were obtained from ATCC and were grown in ATCC-formulated RPMI1640 Medium (cat. 30-2001) with supplements. In some cases, the prostate cancer cells were cultured in RPMI with 1% FBS supplemented with 200 ng/mL of human recombinant CXCL12 (cat. 350-NS, R&D Systems). The human breast cancer cell line, MCF7 cells were kindly provided by Dr. Max Wicha (University of Michigan, Ann Arbor, MI). MCF7 cell line was cultured in DMEM with supplements (Invitrogen). All cell lines were routinely monitored for mycoplasma contamination using the Mycoplasma PCR Detection Kit (cat. MP0035, Sigma), and all cells were authenticated by STR genotyping using the Promega 16 High Sensitivity STR Kit (cat. DC2100, Promega). The STR profiles were compared with databases (DSMZ/ATCC/JCRB/RIKEN).

CXCL12γ overexpression

A CXCL12γ overexpression plasmid vector, pLV-CXCL12γ and control vector, pLV were kindly provided by Dr. Ramirez (Viral Vector Facility, Technical Unit of Gene Targeting, Fundacion CNIC, National Centre for Cardiovascular Research, Madrid, Spain; ref. 29). pLV-CXCL12γ and pLV were packaged with lentivirus at University of Michigan. Lentiviral pLV-CXCL12γ or pLV were infected into prostate cancer cells (PC3, DU145, LNCaP, C42B) and breast cancer cells (MCF7). Infected cells were selected for 7 days in media containing 1 μg/mL puromycin and analyzed by real-time qPCR or immunofluorescence staining.

RNAi

PC3 or DU145 cells at 60% confluence were seeded onto 6-well culture plates. After 24 hours, negative control siRNA (cat. 4390843, Ambion) or CXCR4 siRNA (cat. 4390824, Ambion) with OPTI-MEM (cat. 31985-062, Life Technologies) were transfected into prostate cancer cells using Lipofectamine RNAiMAX (cat. 56532, Life Technologies) according to the manufacturer's instructions. Transfected cells were incubated at 37°C for 72 hours, and the cells were used to various cell assays. Silencing was verified by Western blot analysis.

FACS analysis

For analysis of a CSC phenotype (CD133+/CD44+), overexpression of CXCL12γ in prostate cancer cells or control cells (PC3, DU145; 1 × 105) were seeded onto 12-well culture plates and were cultured for 4 days. The cells were incubated with PE-anti-CD133 antibody (cat. 130-080-901, Miltenyi Biotec) and APC-anti-CD44 antibody (cat. 559942, BD Biosciences) for 20 minutes at 4°C. For CXCR4+ cell analysis, the cells were incubated with PE-anti-human CD184 (CXCR4) antibody (cat. 306506, BioLegend) or mouse IgG-PE (cat. 130-092-212, Miltenyi Biotec) for 20 minutes at 4°C. The CD133+/CD44+ or CXCR4+ fractions were analyzed with a FACS Aria High-Speed Cell Sorter (BD Biosciences). Apoptosis was measured by flow cytometry (FACSAria dual laser flow cytometer, Becton Dickinson) using PE Annexin V Apoptosis Detection Kit I (cat. 559763, BD Biosciences). The prostate cancer cells were pretreated AMD3100 (5 μg/mL) or siCXCR4 and treated with of docetaxel (Taxotere; 0.5–1 μg/mL, Hospira). In some cases, the prostate cancer cells were treated with XTANDI (enzalutamide; 0.5 μg/mL; Selleck Chemicals).

Prostatosphere culture and assay

Prostate cancer cells that overexpress CXCL12γ or control (PC3, DU145) were dissociated to single cells by standard trypsinization and washed three times with PBS. The cells were plated in stem cell culture medium (DMEM:F12 plus 10 ng/mL bFGF, 20 ng/mL EGF, 5 mg/mL insulin, and 0.4% BSA) supplemented with 1% KO serum replacement (cat. 10828-028, Invitrogen/Gibco) at a density of 1,000 cells/mL in low attachment 6-well culture plates (31). Seven-day-old spheres are enumerated as size >50 cells.

Quantitative RT-PCR

Total RNA was extracted from cells using the RNeasy Mini or Micro Kit (Qiagen) and converted into cDNA using a First-Strand Synthesis Kit (Invitrogen). Quantitative PCR was performed on an ABI 7700 sequence detector (Applied Biosystems) using TaqMan Universal PCR Master Mix Kit (Applied Biosystems) according to the directions of the manufacturer. TaqMan MGB probes (Applied Biosystems) were as follows: CXCL12γ (hsSDF-1YJC), CXCR4 (Hs00237052_m1), AR-V7 (Hs04260217_m1), E-cadherin (CDH1; Hs010113953_m1), N-cadherin (CDH2; Hs00169953_m1), vimentin (Hs00185584_m1), α-SMA (Hs00426835_g1), CD44 (Hs01075861_m1), SNAIL1 (Hs00195591_m1), SNAIL2 (Hs0095344_m1), ZEB1 (Hs00232783_m1), ZEB2 (Hs00207691_m1), TGFBR2 (Hs00234253_m1), androgen receptor (AR; Hs00171172_m1), N-MYC (NYCN; 00232074_m1), NDRG1 (Hs00608387_m1), ENO2 (Hs00157360_m1), and synaptophysin (Hs00300531_m1). β-Actin (Hs99999903_m1) was used as internal controls for the normalization of target gene expression.

For the analysis of gene expression for circulating tumor cells (CTC), the healthy normal subjects (n = 2) and patients with m-CRPC (n = 26) were enrolled in the companion of the protocol “Evaluation of blood and bone marrow elements that contribute to genitourinary cancer metastasis” at the University of Michigan Comprehensive Cancer Center after obtaining a separate written informed consent approved by the University of Michigan Institutional Review Board in accordance with the Declaration of Helsinki. The whole blood (5 mL) was drawn from the health normal subjects and prostate cancer patients (IRB: 00052405, University of Michigan). CTCs were isolated from blood using anti-EpCAM conjugated Dynabeads (Thermo Fisher Scientific; refs. 32–34). After 5 times wash with PBSF (PBS plus 0.1% FBS), CTCs were lysed on beads with the lysis buffer and stored at −80°C. mRNA from CTCs was captured using Oligo(dT)25 mRNA Dynabeads (Thermo Fisher Scientific) and reverse transcribed into cDNA. Target gene amplified library was generated with multiplex PCR with a pooled primer set including 150 genes. Real-time PCR was then used to determine individual gene expression level. Blood from healthy normal controls (no history of cancer) were used to establish baseline gene expression related to WBC contamination.

CXCL12γ expression was also analyzed in normal human tissues and C57BL/6 mouse tissues (n = 3 mice). Total human RNA was purchased from Life Technologies; brain (cat. AM 7962), heart (cat. AM7966), lung (cat. AM7968), liver (cat. AM7960), spleen (cat. AM7960), kidney (cat. AM7976), prostate (cat. AM7988), testis (cat. AM7972), and skeletal muscle (cat. AM7982). The human or mouse-specific Cxcl12γ primers were designed through Life Technologies (human CXCL12γ primer, cat. SDF-1YJC; mouse Cxcl12γ primer, cat. CXCL12C). The human CXCL12γ TaqMan probes are F–5′-CCA AGA GTA CCT GGA GAA AGC TTT AA-3′, R–5′-CGG TCC ATC GGC AGG AA-3′, TaqMan probe–5′-FAM-TTC TCT TCT TCT GTC GCT TCT-TAMRA-3′ (Applied Biosystems) and the mouse Cxcl12γ TaqMan probes are F–5′-CCA AGA GTA CCT GGA GAA AGC TTT AA-3′, R–5′-CAC GGA TGT CAG CCT TCC T-3′, TaqMan probe–5′-FAM- AAG TAA GCA CAA CAG CCC-TAMRA-3′ (Applied Biosystems).

IHC

Cells and tumor sections were used for immunostaining. Cells were fixed and permeabilized with Perm/Wash Buffer (cat. 554723, BD Biosciences). Tumor sections were blocked with Image-iT FX signal enhancer for 30 minutes and incubated for 2 hours at room temperature with primary antibodies combined with reagents of Zenon Alexa Fluor 488 (green; cat. Z25002, Invitrogen) or 555 (red; cat. Z25305, Invitrogen) labeling kit. For the CXCL12γ antibody, a rabbit polyclonal antiserum (anti-CXCL12γ) was raised against the peptide KVGKKEKIGKKKRQ, mapping to the specific C-terminal region of CXCL12γ. The N-terminal cysteine enables direct conjugation of the peptide to the protein carrier and is not present in the native sequence. Peptide synthesis, coupling, immunization, ELISA titration, and affinity purification were done by BioGenes GmbH (29). Cxcl12α (mouse, cat. ab25117, Abcam), CD133 (cat. 130-090-423, Miltenyi Biotec), CD44 (cat. ab6124, Abcam), CXCR4 (cat. ab181020, Abcam), CD24 (cat. MS-1279-P, Thermo Fisher Scientific), ENO2 (cat. NB110-58870, Novus), synaptophysin (cat. ab23754, Abcam), pan-cytokeratin (cat. ab86734, Abcam), and α-tubulin (cat. ab52866, Abcam) antibodies were used as primary antibody. After washing with PBS, tumor sections were mounted with ProLong Gold antifade reagent with DAPI (cat. P36931, Invitrogen). Images were taken with Olympus FV-500 confocal microscope. In some cases, hematoxylin and eosin (H&E) stain, ENO2, synaptophysin, pan-cytokeratin antibodies were used for IHC staining in subcutaneous tumors. Images were taken with Olympus 51A microscope. Human prostate tissue microarrays (TMA) were obtained from The Tissue Core of the University of Michigan Comprehensive Cancer Center and US Biomax, Inc. TMAs consist of normal prostate tissues (n = 8), benign (n = 6), Gleason 6–7 prostate cancer tissues (n = 12), Gleason 9-1-0 prostate cancer tissues (n = 25). Tumors were graded using stage progressing system. Human prostate metastatic tissues [lymph node (n = 15), lung (n = 10), liver (n = 12), and bone (n = 19)] also obtained from The Tissue Core of the University of Michigan Comprehensive Cancer Center. Staining intensity was ranked on a scale from 1 to 4 (1, negative; 2, weak; 3, moderate; and 4, strong).

Western blots

Whole-cell lysates were separated on 4% to 20% Tris-Glycine gel and transferred to PVDF membranes. The membranes were incubated with 5% milk for 1 hour and incubated with primary antibodies overnight at 4°C. Primary antibodies used included anti-MARCKS (1:1,000 dilution, cat. 5607, Cell Signaling Technology), anti-p-MARCKS (Ser159/163; 1:1,000 dilution, cat. 11992, Cell Signaling Technology), anti-PKCα (1:1,000 dilution, cat. 2056, Cell Signaling Technology), anti–p-PKCα (1:1,000 dilution, cat. 9373, Cell Signaling Technology), monoclonal anti-NFκBp65, 1:1,000 dilution, cat. 8242, Cell Signaling Technology), monoclonal anti-p-NFκBp65, 1:1,000 dilution, cat. 3033, Cell Signaling Technology), anti-CXCR4 (1:1,000 dilution, cat. ab181020, Abcam), polyclonal anti-N-MYC (1:1,000 dilution, cat. 9405, Cell Signaling Technology), monoclonal anti-NDRG1 (1:1,000 dilution, cat. 9485, Cell Signaling Technology), polyclonal anti-ENO2 (1:1,000 dilution, cat. 2056, Cell Signaling Technology), and polyclonal anti-synaptophysin (1:1,000 dilution, cat. 4329, Cell Signaling Technology). Blots were incubated with peroxidase-coupled anti-mouse IgG secondary antibody (cat. 7076, 1:2,000 dilution, Cell Signaling Technology) or peroxidase-coupled anti-rabbit IgG secondary antibody (cat. 7074, 1:2,000 dilution, Cell Signaling Technology) for 1 hour, and protein expression was detected with SuperSignal West Dura Chemiluminescent Substrate (cat. Prod 34075, Thermo Fisher Scientific). Membranes were reprobed with monoclonal anti–β-actin antibody (1:1,000 dilution; cat. 4970, Cell Signaling Technology) to control for equal loading.

PKC ELISA

PC3 or DU145 cells (1 × 105) were seeded onto 12-well culture plates and cultured for 4 days. Whole-cell lysates were prepared to evaluate PKC kinase activity by following the directions of the manufacturer (cat. ADI-EKS-420A, Enzo Life Sciences). The levels of PKC kinase activity were normalized to total cell numbers.

Enolase 2, chromogenin A, and PSA ELISA

For the analysis of serum enolase 2, chromogenin A, and PSA levels for CTCs from prostate cancer patients, serum samples were collected from whole blood, which were drawn from prostate cancer patients (IRB: 00052405, University of Michigan). Serum enolase 2 and chromogenin A were evaluated following the manufacturer's directions (enolase2/NSP, cat. DENL20, R&D System; chromogenin A, cat. DY9098, R&D Systems). PSA data were provided by University of Michigan Cancer Center (IRB: 00052405, University of Michigan).

NFκB luciferase reporter assay

Dual-luciferase reporter assays (cat. E1910, Dual-Lucuferase Reportor Assay System, Promega) were performed according to the manufacturer's instructions. Cells plated in 24-well plates were transiently transfected with NFκB promoter-firefly luciferase plasmid and internal control plasmid pRL-TK (Promega), which encodes Renilla luciferase and is used to normalize transfection efficacy. The cells were incubated for 48 hours. All firefly luciferase activity was normalized to Renilla luciferase activity or total DNA (cat. P11496, picoQuant-iT PicoGreen dsDNA Assay Kit, Molecular Probes), and data were presented as relative luciferase activity where control values were set to 1 for each cell line.

Subcutaneous tumor growth and in vivo metastasis assay

Tumors were established by injecting prostate cancer cells (CXCL12γ-overexpressing prostate cancer cells or controls) or breast cancer cells (CXCL12γ-overexpressing breast cancer cells or controls; 1 × 106) in growth factor–reduced Collagen Type I gel (cat. 354236, BD Biosciences) subcutaneously into 5- to 7-week-old male or female SCID mice. The animals were monitored daily. Tumor volumes were calculated using the formula V = (the shortest diameter) × (the longest diameter) × height. After 4 weeks, the animals were sacrificed. The tumors were measured and prepared for histology. For in vivo metastasis assays, IHC for prostate cancer cells in the marrow was also used. The numbers of prostate cancer cells were quantified on the endosteal region defined as 10 cell diameters from the bone surfaces.

In vivo isolation of disseminated prostate cancer

The prostate cancer cells overexpressing CXCL12γ or control cells (1 × 106 cells) were injected into male 5- to 7-week-old male SCID mice by intracardiac injection. Groups were PBS (n = 2), PC3-control cells (n = 7), and PC3- CXCL12γ-overexpressing cells (n = 8). After 72 hours, brain, lung, liver, and bone (spine, pelvis, femur, and tibia) tissues were collected. The cells from animal tissues were incubated first with a RBC lysis buffer (Qiagen, cat. 8305333), and then were incubated with a PE/Cy7 anti-mouse H-2Kd (MHC) antibody (cat. 116622, BioLegend) and APC/Cy7 anti-human HLA-ABC antibody (cat. 311426, BioLegend) for 30 minutes at 4°C. The disseminated prostate tumor cells (prostate cancer DTC) were analyzed with a FACSAria II Cell Sorter by gating on mouse MHC-negative and HLA-ABC–positive cells. Data are presented as mean ± SD (Student t test).

Statistical analyses

Results are presented as mean ± SD. Significance of the difference between two measurements was determined by unpaired Student t test. Correlation was analyzed with simple linear regression to calculate Pearson correlation coefficient (r) and P value. Analyses were conducted in with GraphPad Prism version 7 software. Values of P < 0.05 were considered statistically significant.

CXCL12γ is associated with malignancy in prostate cancer

We previously reported that elevated expression of CXCL12 and its receptor CXCR4 are found in prostate cancer tissues and cell lines and are associated with increasing malignant potential (22). Here, the role of the alternatively transcribed CXCL12 isoform CXCL12γ, an intracellular chemokine, was explored in relation to prostate tumor progression. Using human prostate tumor and metastatic TMAs, CXCL12γ expression was examined using a polyclonal antibody raised against a peptide specific to the C-terminal region of CXCL12γ. Little or no expression of CXCL12γ was detected in normal prostate and benign prostate cancer tissues, and relatively low levels of CXCL12γ expression were observed in low-grade prostate cancer tissues and significant expression of CXCL12γ was detected in high-grade prostate cancer tissues dominated by small-cell carcinoma (Fig. 1A and B). In the metastatic tissues from m-CRPC patients, significant CXCL12γ-expressing small-cell carcinoma was detected in lymph node, lung, liver, and bone tissues (Fig. 1C and D). In contrast, CXCL12γ expression was not detected in regions dominated by luminal type of adenocarcinoma cells from the high-grade prostate cancer tissues (Fig. 1A; Supplementary Fig. S1A and S1B). CXCL12γ expression in bone metastases from m-CRPC patients mirrored that of the primary lesions; CXCL12γ expression was observed in epithelial regions (cytokeratin-positive) dominated by a small-cell phenotype, whereas it was not observed in regions dominated by luminal type of adenocarcinoma cells (Fig. 1E; Supplementary Fig. S1C and S1D).

Figure 1.

CXCL12γ expression in prostate cancer patient tissues and m-CRPC cells. A, Human prostate TMAs from The Tissue Core of the University of Michigan Comprehensive Cancer Center and US Biomax Inc. were stained for CXCL12γ (red) or pan-cytokeratin (green). Cells expressing both markers are indicated with white arrows. DAPI nuclear stain (blue). Scale bar, 50 μm. TMAs consist of normal prostate tissues (n = 8), benign (n = 6), Gleason 6–7 prostate cancer tissues (n = 12), Gleason 9–10 prostate cancer tissues (n = 25). B, Quantification of A. C, Human prostate metastatic tissues from The Tissue Core of the University of Michigan Comprehensive Cancer Center were stained for CXCL12γ (red) or pan-cytokeratin (green). Metastatic tissues are lymph node (n = 15), lung (n = 10), liver (n = 12), and bone (N = 19). D, Quantification of C. E, Left, CXCL12γ (red) expression in pan-cytokeratin (green) expressing prostate adenocarcinoma in human bone tissue as detected by immunofluorescence staining, and the magnification of the yellow rectangle from left panel. Scale bar, 20–50 μm. Right, CXCL12γ (red) expression in pan-cytokeratin (green) expressing a small-cell carcinoma phenotype in human bone tissue as detected by immunofluorescence staining, and the magnification of the yellow rectangle from right panel (white arrows). Scale bar, 20–50 μm. F, mRNA expression of CXCL12γ in CSCs (CD133+/CD44+) or non-CSCs (CD133/CD44) from LNCaP cells or a castration-resistant subline of the LNCaP cells, LNCaP95 as quantified by real-time PCR. G, mRNA expression of CXCL12γ in CSCs (CD133+/CD44+) or non-CSCs (CD133/CD44) from the metastatic cell lines PC3 and DU145 as quantified by real-time PCR. Data in B, D, F, and G are representative of mean ± SD (Student t test).

Figure 1.

CXCL12γ expression in prostate cancer patient tissues and m-CRPC cells. A, Human prostate TMAs from The Tissue Core of the University of Michigan Comprehensive Cancer Center and US Biomax Inc. were stained for CXCL12γ (red) or pan-cytokeratin (green). Cells expressing both markers are indicated with white arrows. DAPI nuclear stain (blue). Scale bar, 50 μm. TMAs consist of normal prostate tissues (n = 8), benign (n = 6), Gleason 6–7 prostate cancer tissues (n = 12), Gleason 9–10 prostate cancer tissues (n = 25). B, Quantification of A. C, Human prostate metastatic tissues from The Tissue Core of the University of Michigan Comprehensive Cancer Center were stained for CXCL12γ (red) or pan-cytokeratin (green). Metastatic tissues are lymph node (n = 15), lung (n = 10), liver (n = 12), and bone (N = 19). D, Quantification of C. E, Left, CXCL12γ (red) expression in pan-cytokeratin (green) expressing prostate adenocarcinoma in human bone tissue as detected by immunofluorescence staining, and the magnification of the yellow rectangle from left panel. Scale bar, 20–50 μm. Right, CXCL12γ (red) expression in pan-cytokeratin (green) expressing a small-cell carcinoma phenotype in human bone tissue as detected by immunofluorescence staining, and the magnification of the yellow rectangle from right panel (white arrows). Scale bar, 20–50 μm. F, mRNA expression of CXCL12γ in CSCs (CD133+/CD44+) or non-CSCs (CD133/CD44) from LNCaP cells or a castration-resistant subline of the LNCaP cells, LNCaP95 as quantified by real-time PCR. G, mRNA expression of CXCL12γ in CSCs (CD133+/CD44+) or non-CSCs (CD133/CD44) from the metastatic cell lines PC3 and DU145 as quantified by real-time PCR. Data in B, D, F, and G are representative of mean ± SD (Student t test).

Close modal

The intriguing observation that CXCL12γ expression is associated with progressive disease and with cellular regions dominated by a small-cell phenotype raises the possibility that CXCL12γ may serve as a marker of CSCs in m-CRPC. To address these possibilities, cells enriched for CSC-like activities were isolated from prostate cancer cell lines based upon their expression of CD133 and CD44 (14) and examined for CXCL12γ expression. CXCL12γ expression in both hormone-sensitive and castration-resistant CSCs was significantly enhanced relative to non-CSCs (Fig. 1F). Importantly, even among the CSC populations, CXCL12γ expression was significantly enhanced in castration-resistant cell line LNCaP95 compared with the hormone-sensitive cell line LNCaP (Fig. 1F). CXCL12γ expression was also significantly enhanced in CSCs relative to non-CSCs from the metastatic cell lines PC3 and DU145 (Fig. 1G). These data suggest that CXCL12γ expression is associated with the CSC phenotype and m-CRPC.

CXCL12γ induces CSCs, which promote prostate tumor growth and metastasis

Under normal culture conditions, little or no expression of CXCL12γ was detected in human prostate cancer cell lines (Supplementary Fig. S2A). To further explore the role that CXCL12γ plays in tumor progression, CXCL12γ was overexpressed in prostate cancer (PC3 and DU145). Overexpression of CXCL12γ was confirmed at the RNA and protein levels (Fig. 2A and B). As higher levels of CXCL12γ expression were observed in the CSC population in castration-resistant prostate cancer cells in Fig. 1F, studies were performed to evaluate the impact of CXCL12γ on the CSC phenotype in vitro. Overexpression of CXCL12γ led to a significant increase in the expression of the CSC phenotype (Fig. 2C) and facilitated the development of spheres in culture (Fig. 2D). To validate that CXCL12γ plays an active role in tumor growth, prostate cancer cells were implanted subcutaneously into SCID mice and evaluated for tumor growth. Intriguingly, tumors generated from the CXCL12γ-overexpressing prostate cancer cells grew significantly larger than tumors generated from the control cells (Fig. 2E–G). We further confirmed that significant numbers of CSCs (CD133+/CD44+ cells) in prostate cancer tumors were detected in tumors generated with CXCL12γ-overexpressing cancer cells compared with control cells (Fig. 2H and I). Consistent with more aggressive and larger tumor growth observed following subcutaneous injection, enhanced numbers of disseminated tumor cells (DTC) were observed in bone tissues of these animals following injection of CXCL12γ overexpressing prostate cancer cells compared with control cells (Fig. 2J and K). The enhanced number of DTCs found in the bone marrows of these animals was likely made possible by the induction of an EMT in cells overexpressing CXCL12γ (Supplementary Fig. S2B). To further validate that CXCL12γ plays an active role in tumor metastasis, the prostate cancer cells overexpressing CXCL12γ or control cells were injected intracardially into SCID mice and evaluated for DTCs. Significant numbers of prostate cancer DTCs were observed from PC3 CXCL12γ-overexpressing cells compared with PC3-control cells in brain, lung, liver, and bone tissues (Fig. 2L; Supplementary Fig. S2C). In addition, we also observed that high levels of CXCL12γ expression are present in normal brain, lung, liver, and heart, adrenal gland, and bone tissues (Supplementary Fig. S2D and S2E). Together, these data suggest that CXCL12γ induces a CSC phenotype, which promotes prostate cancer tumor outgrowth and ultimately enhanced levels of DTCs in the soft and bone tissues. To further validate that CXCL12γ plays an active role in tumor metastasis, the prostate cancer cells overexpressing CXCL12γ or control cells were injected intracardially into SCID mice and evaluated for DTCs. Significant numbers of prostate cancer DTCs were observed from PC3- CXCL12γ-overexpressing cells compared with PC3-control cells in brain, lung, liver, and bone tissues (Fig. 2L; Supplementary Fig. S2C). In addition, we also observed that high levels of CXCL12γ expression are present in normal brain, lung, liver, heart, adrenal gland, and bone tissues (Supplementary Fig. S2D and S2E).

Figure 2.

CXCL12γ induces CSCs, which promotes prostate tumor growth and metastasis. A, Verification of CXCL12γ mRNA expression in CXCL12γ-overexpressing prostate cancer cells, PC3 and DU145, as quantified by real-time PCR. B, Verification of CXCL12γ protein expression in CXCL12γ-overexpressing prostate cancer cells, PC3 and DU145, as detected by immunofluorescence staining. CXCL12γ, green; CD44, red. DAPI nuclear stain, blue. Scale bar, 20 μm. C, FACS analyses of the CSC (CD133+/CD44+) phenotype in CXCL12γ-overexpressing or control prostate cancer cells. D, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells. Data in A, C, and D are presented as mean ± SD (Student t test). E–G, The GFP-expressing prostate cancer cells overexpressing CXCL12γ or control cells were implanted subcutaneously into 5- to 7-week-old male SCID mice (n = 8/group). After 4 weeks, mice were sacrificed and tumor growth was evaluated. *, P < 0.001. H, Immunostaining of CD133 (green) and CD44 (red; white arrow) of subcutaneous prostate cancer tumors generated by CXCL12γ-overexpressing prostate cancer cells or control cells. I, Quantification of H. Data, mean ± SD (Student t test). J, GFP-expressing prostate cancer cells were identified in the femur of SCID mice following subcutaneous injection in E. Green arrows, GFP-expressing prostate cancer cells. Red arrows, osteoblast on the bone surface staining positive for CXCL12 expression. Scale bar, 50 μm. K, Quantification of J. The numbers of GFP-expressing prostate cancer cells were quantified on the endosteal region of the 10 long bones. Endosteal regions were defined as 12 cell diameters from bone surfaces. Mean ± SD (Student t test; n = 10). L, Prostate cancer cells overexpressing CXCL12γ or control cells (1 × 106 cells) were injected into male CB.17. SCID mice (5–7 weeks of age; Charles River Laboratories) by intracardiac injection. Groups were PBS (n = 2), PC3 control cells (n = 7), and PC3 CXCL12γ-overexpressing cells (n = 8). Disseminated prostate tumor cell numbers (prostate cancer DTC) were analyzed on a FACSAria II Cell Sorter by gating on mouse MHC-negative and HLA-ABC–positive cells from brain, lung, liver, and bone tissues 72 hours later. Data, mean ± SD (Student t test).

Figure 2.

CXCL12γ induces CSCs, which promotes prostate tumor growth and metastasis. A, Verification of CXCL12γ mRNA expression in CXCL12γ-overexpressing prostate cancer cells, PC3 and DU145, as quantified by real-time PCR. B, Verification of CXCL12γ protein expression in CXCL12γ-overexpressing prostate cancer cells, PC3 and DU145, as detected by immunofluorescence staining. CXCL12γ, green; CD44, red. DAPI nuclear stain, blue. Scale bar, 20 μm. C, FACS analyses of the CSC (CD133+/CD44+) phenotype in CXCL12γ-overexpressing or control prostate cancer cells. D, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells. Data in A, C, and D are presented as mean ± SD (Student t test). E–G, The GFP-expressing prostate cancer cells overexpressing CXCL12γ or control cells were implanted subcutaneously into 5- to 7-week-old male SCID mice (n = 8/group). After 4 weeks, mice were sacrificed and tumor growth was evaluated. *, P < 0.001. H, Immunostaining of CD133 (green) and CD44 (red; white arrow) of subcutaneous prostate cancer tumors generated by CXCL12γ-overexpressing prostate cancer cells or control cells. I, Quantification of H. Data, mean ± SD (Student t test). J, GFP-expressing prostate cancer cells were identified in the femur of SCID mice following subcutaneous injection in E. Green arrows, GFP-expressing prostate cancer cells. Red arrows, osteoblast on the bone surface staining positive for CXCL12 expression. Scale bar, 50 μm. K, Quantification of J. The numbers of GFP-expressing prostate cancer cells were quantified on the endosteal region of the 10 long bones. Endosteal regions were defined as 12 cell diameters from bone surfaces. Mean ± SD (Student t test; n = 10). L, Prostate cancer cells overexpressing CXCL12γ or control cells (1 × 106 cells) were injected into male CB.17. SCID mice (5–7 weeks of age; Charles River Laboratories) by intracardiac injection. Groups were PBS (n = 2), PC3 control cells (n = 7), and PC3 CXCL12γ-overexpressing cells (n = 8). Disseminated prostate tumor cell numbers (prostate cancer DTC) were analyzed on a FACSAria II Cell Sorter by gating on mouse MHC-negative and HLA-ABC–positive cells from brain, lung, liver, and bone tissues 72 hours later. Data, mean ± SD (Student t test).

Close modal

To further explore the role that CXCL12γ plays in tumor progression, CXCL12γ was overexpressed in breast cancer cell line (MCF7). Under normal culture conditions, little or no expression of CXCL12γ was detected in human breast cancer cell line MCF7 (Supplementary Fig. S3A). Overexpression of CXCL12γ was confirmed at the RNA and protein levels (Supplementary Fig. S3A and S3B). Overexpression of CXCL12γ led to the development of spheres in culture (Supplementary Fig. S3C and S3D). To validate that CXCL12γ plays the role in tumor growth, breast cancer cells were implanted subcutaneously into SCID mice and evaluated for tumor growth. The tumors generated from the CXCL12γ-overexpressing breast cancer cells grew significantly larger than tumors generated from the control cells (Supplementary Fig. S3E–S3G). We further confirmed that significant numbers of CSCs (CD24/CD44+ cells) in breast cancer tumors were detected in tumors generated with CXCL12γ-overexpressing cancer cells compared with control cells (Supplementary Fig. S3H and S3I). We also found that breast cancer cells overexpressing CXCL12γ increase the induction of EMT markers, suggesting the implication of metastatic potentials (Supplementary Fig. S3J).

Together, these data suggest that CXCL12γ induces a CSC phenotype that promotes tumor outgrowth and ultimately enhanced levels of DTCs in the soft and bone tissues.

CXCL12γ induces neuroendocrine phenotype, which promotes the development of m-CRPC

Neuroendocrine prostate cancer disease is commonly observed in relapsing individuals particularly after treatment with second-generation antiandrogens (e.g., abiraterone and enzalutamide; refs. 10, 12, 14, 35–38). Emerging evidence indicates that the neuroendocrine cell differentiation and more aggressive disease are associated with enhanced expression of CSC and EMT phenotypes (14, 35, 36). To explore the extent to which CXCL12γ expression is associated with neuroendocrine differentiation, we examined the expression of a neuroendocrine phenotype in CXCL12γ-overexpressing prostate cancer cells. In vitro, neuroendocrine markers were significantly increased in CXCL12γ overexpressing prostate cancer cells compared with control cells (Fig. 3A and B). To validate the in vitro studies, enolase-2 (ENO2) and synaptophysin staining were performed on the tumors generated with CXCL12γ-overexpressing prostate cancer cells compared with control cells. A higher density of neuroendocrine cells with extensive vascularization (Fig. 3C and D, H&E stain), ENO2, and synaptophysin expression were observed in tumors generated from the cells overexpressing CXCL12γ compared with control cells (Fig. 3C and D). Pan-cytokeratin expression was used to confirm that these neuroendocrine prostate cancer cells were of human origin (Fig. 3C and D). To further explore the extent to which CXCL12γ expression is associated with neuroendocrine differentiation in breast cancer cell line (MCF7), we examined the expression of a neuroendocrine phenotype in CXCL12γ-overexpressing breast cancer cells or control cells. In vitro, neuroendocrine markers were significantly increased in CXCL12γ-overexpressing breast cancer cells compared with control cells (Supplementary Fig. S4A). We also examined the expression of neuroendocrine markers in breast cancer tumors. A higher density of neuroendocrine cells (Supplementary Fig. S4B, H&E stain) with ENO2 and synaptophysin expression were observed in tumors generated from the cells overexpressing CXCL12γ compared with control cells (Supplementary Fig. S4B). Pan-cytokeratin expression was used to confirm that these neuroendocrine cells were of human origin (Supplementary Fig. S4B).

Figure 3.

CXCL12γ induces a neuroendocrine phenotype in prostate cancer and CXCL12γ detection in CTCs of prostate cancer patients. A, mRNA levels of N-MYC (MYCN), N-MYC downstream regulated 1 (NDRG1), enolase-2 (ENO2), and synaptophysin (SYP) expression following CXCL12γ-overexpressing or control prostate cancer cells as quantified by real-time PCR. B, Protein levels of N-MYC, NDRG1, ENO2, and synaptophysin in CXCL12γ-overexpressing or control prostate cancer cells as quantified by Western blots. Examination of neuroendocrine markers in subcutaneous tumors generated by CXCL12γ-overexpressing or control prostate cancer cells in SCID mice in Fig. 2E. High density of neuroendocrine cells with extensive vascularization in PC3 tumors (C) and DU145 tumors (D) is shown by H&E staining. Scale bar, 50 μm. ENO2-, synaptophysin-, and pan-cytokeratin–expressing neuroendocrine cells as detected by IHC staining. Scale bar, 50 μm. E, Gene expression of AR-V7, CXCL12γ, and ENO2 in CTCs isolated from healthy normal control (n = 2) and m-CRPC patients (n = 26) as quantified by real-time PCR. F, Regression analysis between ENO2 and CXCL12γ. Data, mean ± SD (Student t test).

Figure 3.

CXCL12γ induces a neuroendocrine phenotype in prostate cancer and CXCL12γ detection in CTCs of prostate cancer patients. A, mRNA levels of N-MYC (MYCN), N-MYC downstream regulated 1 (NDRG1), enolase-2 (ENO2), and synaptophysin (SYP) expression following CXCL12γ-overexpressing or control prostate cancer cells as quantified by real-time PCR. B, Protein levels of N-MYC, NDRG1, ENO2, and synaptophysin in CXCL12γ-overexpressing or control prostate cancer cells as quantified by Western blots. Examination of neuroendocrine markers in subcutaneous tumors generated by CXCL12γ-overexpressing or control prostate cancer cells in SCID mice in Fig. 2E. High density of neuroendocrine cells with extensive vascularization in PC3 tumors (C) and DU145 tumors (D) is shown by H&E staining. Scale bar, 50 μm. ENO2-, synaptophysin-, and pan-cytokeratin–expressing neuroendocrine cells as detected by IHC staining. Scale bar, 50 μm. E, Gene expression of AR-V7, CXCL12γ, and ENO2 in CTCs isolated from healthy normal control (n = 2) and m-CRPC patients (n = 26) as quantified by real-time PCR. F, Regression analysis between ENO2 and CXCL12γ. Data, mean ± SD (Student t test).

Close modal

Detection of CTCs in blood is a potential prognostic marker in m-CRPC patients and has also been used as a tool to assess gene expression of metastatic tumors (4–6, 12). For example, AR-V7–based CTC detection has been shown to be an accurate predictive marker of response to enzalutamide and abiraterone in patients with m-CRPC (4–6). Here, we measured expression of CXCL12γ and the neuroendocrine marker ENO2 in CTCs of m-CRPC patients to assess the association of CXCL12γ with neuroendocrine disease (Fig. 3E). Critically, we observed that CXCL12γ expression was significantly higher in men with increased ENO2 expression (P = 0.018), suggesting that CXCL12γ is associated with a neuroendocrine state (Fig. 3F; ref. 10). In addition, we observed a trend of positive linear relationship in ENO2 mRNA expression in CTCs and serum enolase and chromogranin A (P = 0.129 and P = 0.131, respectively) and an inverse linear relationship (P = 0.250) in m-CRPC patients (Supplementary Fig. S5A–S5C). Together, these data suggest that CXCL12γ induces an aggressive neuroendocrine phenotype, which contributes to the development of m-CRPC.

CXCL12γ contributes to the activation of CXCR4 signaling in CSCs and neuroendocrine inductions and resistance in prostate cancer

To determine the impact of the major CXCL12 receptor, CXCR4, in CSC activities and resistance, CSCs were examined from hormone-sensitive cell line or castration-resistant cell line and CXCR4 expression was also examined in CSCs or non-CSCs from hormone-sensitive cell line or castration-resistant cell line. CSCs were significantly elevated in the castration-resistant LNCaP95 cells compared with the parental hormone-sensitive LNCaP cells (Fig. 4A) and neuroendocrine prostate cancer cells (NCI-H660; Supplementary Fig. S6A and S6B). CXCR4 expression in castration-resistant CSCs was significantly enhanced relative to hormone-sensitive CSCs (Fig. 4B). Total CXCR4 protein levels were enhanced in castration-resistant LNCaP95 cells compared with the parental hormone-sensitive LNCaP cells (Fig. 4C). These data suggest that CXCL12γ signaling is linked to induction of a CSC phenotype possibly through its receptor, CXCR4. We further verified that the expression levels of CXCL12γ and CXCR4 were dramatically enhanced in cells with a neuroendocrine phenotype (NCI-H660) compared with PC3 and DU145 cells (Fig. 4D and E). Critically, colocalization of CXCL12γ with CXCR4 near the nuclear membrane was observed in NCI-H660 cells (Fig. 4F). We next probed the cells for expression of CXCR4, which is strongly linked to metastasis in solid tumors, including prostate cancer (23). Overexpression of CXCL12γ significantly increased the expression of CXCR4 in prostate cancer cells (Fig. 4G). Critically, intracellular CXCR4 colocalized with CXCL12γ near the nucleus of CXCL12γ-expressing prostate cancer cells in a similar pattern as in neuroendocrine prostate cancer cells (NCI-H660; Fig. 4H; Supplementary Fig. S6C). To further examine CXCR4 involvement in CXCL12γ-mediated CSC induction, the prostatosphere formation assays were performed in the presence or absence of the small-molecule inhibitor of CXCR4, AMD3100. AMD3100 inhibited the formation of the prostatospheres in control cells, but had minimal impact on the prostatosphere formation in cells overexpressing CXCL12γ, further suggesting that intracellular CXCR4 is more involved in CSC activities than the membrane-binding CXCR4 (Fig. 4I and J). Consistent with these observations, blocking CXCR4 signaling with AMD3100 sensitizes prostate cancer cells to docetaxel chemotherapy. However, when cells were induced to overexpress CXCL12γ, AMD3100 did not sensitize prostate cancer cells to docetaxel chemotherapy (Fig. 4K). We also examined the effect of CXCL12γ/CXCR4 signaling on docetaxel-induced microtubule stabilization. CXCL12γ/CXCR4 signaling significantly reduced docetaxel-induced microtubule stabilization in CXCL12γ-overexpressing prostate cancer cells compared with control prostate cancer cells (Fig. 4L).

Figure 4.

CXCL12γ contributes activation of CXCR4 signaling in CSC and neuroendocrine inductions and resistance in prostate cancer. A, FACS analyses of CSCs (CD133+/CD44+) phenotype in LNCaP or LNCaP95 cells. B, mRNA expression of CXCR4 in CSCs (CD133+/CD44+) or non-CSCs (CD133/CD44) from LNCaP cells or LNCaP95 cells as quantified by real-time PCR. C, FACS analyses of CXCR4 in LNCaP or LNCaP95 cells. Data in A–C are presented as mean ± SD (Student t test). D, Basal levels of CXCL12γ mRNA expression in neuroendocrine cells (NE; NCI-H660) in comparison with PC3 and DU145 cells as quantified by real-time PCR. E, FACS analyses of CXCR4 in neuroendocrine cells in comparison with PC3 and DU145 cells. Data in A–E are presented as mean ± SD (Student t test). F, Immunostaining of CXCL12γ (green) with CXCR4 (red) in neuroendocrine cells. DAPI nuclear stain, blue. Scale bar, 20 μm. G, FACS analyses of percent CXCR4 in CXCL12γ-overexpressing prostate cancer cells or control cells. H, Immunostaining of CXCL12γ (green) and CXCR4 (red) in CXCL12γ-overexpressing or control prostate cancer cells. DAPI nuclear stain, blue. Scale bar, 20 μm. I, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells following the small-molecule inhibitor of CXCR4, AMD3100, treatments as shown by bright-field microscopy. Scale bar, 50 μm. J, Quantification of I. K, FACS analyses of percent apoptosis of CXCL12γ-overexpressing or control prostate cancer cells following docetaxel (DOX) and/or AMD3100 treatments. Data, mean ± SD (Student t test). *, P < 0.001; +, P = 0.001–0.01; #, P = 0.01–0.05. L, Immunostaining analyses of tubulin stabilization in CXCR4 knockdown CXCL12γ-overexpressing prostate cancer cells (with siControl or siCXCR4 for 72 hours) or control prostate cancer cells following docetaxel (DOX; 1 μg/mL) treatment for 24 hours. α-Tubulin, green. DAPI nuclear stain, blue. Scale bar, 20 μm.

Figure 4.

CXCL12γ contributes activation of CXCR4 signaling in CSC and neuroendocrine inductions and resistance in prostate cancer. A, FACS analyses of CSCs (CD133+/CD44+) phenotype in LNCaP or LNCaP95 cells. B, mRNA expression of CXCR4 in CSCs (CD133+/CD44+) or non-CSCs (CD133/CD44) from LNCaP cells or LNCaP95 cells as quantified by real-time PCR. C, FACS analyses of CXCR4 in LNCaP or LNCaP95 cells. Data in A–C are presented as mean ± SD (Student t test). D, Basal levels of CXCL12γ mRNA expression in neuroendocrine cells (NE; NCI-H660) in comparison with PC3 and DU145 cells as quantified by real-time PCR. E, FACS analyses of CXCR4 in neuroendocrine cells in comparison with PC3 and DU145 cells. Data in A–E are presented as mean ± SD (Student t test). F, Immunostaining of CXCL12γ (green) with CXCR4 (red) in neuroendocrine cells. DAPI nuclear stain, blue. Scale bar, 20 μm. G, FACS analyses of percent CXCR4 in CXCL12γ-overexpressing prostate cancer cells or control cells. H, Immunostaining of CXCL12γ (green) and CXCR4 (red) in CXCL12γ-overexpressing or control prostate cancer cells. DAPI nuclear stain, blue. Scale bar, 20 μm. I, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells following the small-molecule inhibitor of CXCR4, AMD3100, treatments as shown by bright-field microscopy. Scale bar, 50 μm. J, Quantification of I. K, FACS analyses of percent apoptosis of CXCL12γ-overexpressing or control prostate cancer cells following docetaxel (DOX) and/or AMD3100 treatments. Data, mean ± SD (Student t test). *, P < 0.001; +, P = 0.001–0.01; #, P = 0.01–0.05. L, Immunostaining analyses of tubulin stabilization in CXCR4 knockdown CXCL12γ-overexpressing prostate cancer cells (with siControl or siCXCR4 for 72 hours) or control prostate cancer cells following docetaxel (DOX; 1 μg/mL) treatment for 24 hours. α-Tubulin, green. DAPI nuclear stain, blue. Scale bar, 20 μm.

Close modal

Furthermore, to determine the role of CXCL12γ in AR-dependent prostate cancer cells, CXCL12γ was overexpressed in LNCaP and C42B cells (Supplementary Fig. S6D and S6E). The AR mRNA was enhanced when CXCL12γ was overexpressed in LNCaP and C42B cells (Supplementary Fig. S6F). We also observed that less apoptotic cells in CXCL12γ-overexpressing LNCaP cells compared with control cells following enzalutamide treatment and in CXCL12γ-overexpressing C42B cells compared with control cells following docetaxel treatment (Supplementary Fig. S6G and S6H). We further found that neuroendocrine prostate cancer cells (NCI-H660) are highly resistant to docetaxel treatment (Supplementary Fig. S6I). We also found a significant enhancement of CXCR4 expression in breast cancer cell overexpression of CXCL12γ compared with control cells (Supplementary Fig. S7A). Together, these data suggest that the intracellular CXCL12γ and CXCR4 interactions associate with the induction of a CSC phenotype and play a significant role in the regulation of resistance to chemotherapy.

CXCL12γ contributes to the activation of CXCR4-mediated PKCα/NFκB signaling in CSC and neuroendocrine inductions and resistance in prostate cancer

To identify the molecular mechanisms that are functionally associated with CXCL12γ-mediated induction of CSC and neuroendocrine phenotypes, PKCα/NFκB signaling was explored. We found that PKCα and NFκB signaling were preferentially activated in prostate cancer cells overexpressing CXCL12γ compared with control cells (Fig. 5A–D). To prove that PKCα/NFκB activation is responsible for the enhancement of CSC activities, a pan-PKC inhibitor (Go6983) or a NFκB inhibitor (IKK2 inh) was employed. Using each of these inhibitors, significant reductions in CSC phenotype and sphere formation were observed (Fig. 5E–I). To further prove the role of CXCR4 in the activation of NFκB signaling, we examined the phosphorylation of the NFκB subunit p65 following AMD3100 treatment. AMD3100 inhibited CXCL12-mediated NFκB-p65 phosphorylation in parental prostate cancer cells, yet it had little to no impact on NFκB-p65 phosphorylation on the CXCL12γ-overexpressing prostate cancer cells (Fig. 5J and K). To further prove how CXCL12γ/CXCR4–mediated PKCα/NFκB signaling is functionally associated with the development of chemoresistance, myristoylated alanine-rich C-kinase substrate (MARCKS) signaling was explored. Activation of PKC signaling modulates microtubule cytoskeleton and cell morphology (39–41). MARCKS as a downstream target of PKC signaling pathway is known to be critical for regulating multiple pathophysiologic processes, including chemoresistance (40–42). Recent evidence also shows that docetaxel reduces PKC activity targeting of taxol-mediated suppression of NFκB (41, 42). Here, we found that phosphorylation of the MARCKS was preferentially increased in prostate cancer cells overexpressing CXCL12γ (Fig. 5L) and phosphorylation of the MARCKS was significantly reduced along with PKCα and NFκB-p65 phosphorylation in CXCR4 knockdown prostate cancer cells overexpressing CXCL12γ (Fig. 5M). Similarly, we found that significant levels of PKCα and NFκB signaling were activated in breast cancer cells overexpressing CXCL12γ compared with control cells (Supplementary Fig. S7B and S7C).

Figure 5.

CXCL12γ contributes to the activation of CXCR4-mediated PKCα/NFκB signaling in CSC and neuroendocrine inductions and resistance in prostate cancer. A, Levels of total PKC activity in CXCL12γ-overexpressing prostate cancer cells relative to control cells. B, Effect of CXCL12γ on NFκB-reporter luciferase activity. Firefly luciferase was normalized against dsDNA. C, FACS analyses of the CSC (CD133+/CD44+) phenotype in CXCL12γ-overexpressing or control prostate cancer cells following NFκB inhibitor (IKK-2 inhibitor VI) treatments. Data in A–C are representative of mean ± SD (Student t test). D, Immunoblot analysis showing PKCα and its phosphorylation in CXCL12γ-overexpressing or control prostate cancer cells as quantified by Western blots. E and F, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells following the pan-PKC inhibitor (Go6983) treatments as shown by brightfield microscopy (E) and quantification of E (F). G, Immunoblot analysis showing NFκB-p65 and its phosphorylation in CXCL12γ-overexpressing or control prostate cancer cells as quantified by Western blots. H and I, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells following treatment with an NFκB inhibitor (IKK2 inhibitor VI, IKK2 inh) as shown by brightfield microscopy (H) and quantification of H (I). Data in F and I are representative of mean ± SD (Student t test). J and K, Immunoblot analysis showing NFκB-p65 and its phosphorylation in prostate cancer cells following the CXCL12 and/or AMD3100 treatments as quantified by Western blots (J) and CXCL12γ-overexpressing or control prostate cancer cells following AMD3100 treatments (K). L, Immunoblot analysis showing phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS) at Ser159/163 (phospho-MARCKS) in CXCL12γ-overexpressing prostate cancer cells or control prostate cancer cells. M, Immunoblot analysis showing reduction of phosphorylation of p-MARCKS, p-PKCα, and p-NFkB-p65 in CXCR4 knockdown CXCL12γ-overexpressing prostate cancer cells.

Figure 5.

CXCL12γ contributes to the activation of CXCR4-mediated PKCα/NFκB signaling in CSC and neuroendocrine inductions and resistance in prostate cancer. A, Levels of total PKC activity in CXCL12γ-overexpressing prostate cancer cells relative to control cells. B, Effect of CXCL12γ on NFκB-reporter luciferase activity. Firefly luciferase was normalized against dsDNA. C, FACS analyses of the CSC (CD133+/CD44+) phenotype in CXCL12γ-overexpressing or control prostate cancer cells following NFκB inhibitor (IKK-2 inhibitor VI) treatments. Data in A–C are representative of mean ± SD (Student t test). D, Immunoblot analysis showing PKCα and its phosphorylation in CXCL12γ-overexpressing or control prostate cancer cells as quantified by Western blots. E and F, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells following the pan-PKC inhibitor (Go6983) treatments as shown by brightfield microscopy (E) and quantification of E (F). G, Immunoblot analysis showing NFκB-p65 and its phosphorylation in CXCL12γ-overexpressing or control prostate cancer cells as quantified by Western blots. H and I, The formation of prostatospheres in CXCL12γ-overexpressing or control prostate cancer cells following treatment with an NFκB inhibitor (IKK2 inhibitor VI, IKK2 inh) as shown by brightfield microscopy (H) and quantification of H (I). Data in F and I are representative of mean ± SD (Student t test). J and K, Immunoblot analysis showing NFκB-p65 and its phosphorylation in prostate cancer cells following the CXCL12 and/or AMD3100 treatments as quantified by Western blots (J) and CXCL12γ-overexpressing or control prostate cancer cells following AMD3100 treatments (K). L, Immunoblot analysis showing phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS) at Ser159/163 (phospho-MARCKS) in CXCL12γ-overexpressing prostate cancer cells or control prostate cancer cells. M, Immunoblot analysis showing reduction of phosphorylation of p-MARCKS, p-PKCα, and p-NFkB-p65 in CXCR4 knockdown CXCL12γ-overexpressing prostate cancer cells.

Close modal

These data suggest that the intracellular CXCL12γ and CXCR4 interactions are associated with expression of a CSC phenotype and resistance to chemotherapy through PKCα/NFκB signaling pathways.

Our studies demonstrate that CXCL12γ expression in prostate cancer regulates induction of CSC and neuroendocrine phenotypes, promoting prostate cancer tumor outgrowth, metastasis, and chemoresistance, which support the significance impacts of CXCL12γ on the development of m-CRPC.

Despite many advances in therapy for m-CRPC, improvements in survival have generally been on the order of months rather than years (1–7). One possible explanation for this observation is the emergence of metastatic tumor clones or cells with tumor-initiating potential or CSCs, which are extraordinarily resistant to conventional therapy (8–11). This biologic process is mediated by therapy-initiated factors and pathways in the tumor. The second-generation antiandrogen therapies alter intrinsic factors and signaling pathways, including AR, PI3K/AKT/PTEN, HIF, IGF-1, WNT, IL6, SOX2, NANOG, and many others within tumor cells, which facilitate CSC expansion and subsequent m-CRPC development (11). Furthermore, CSCs are associated in the transition from androgen-sensitive to castration-resistant phenotype. For example, PKCα/NFκB signaling is implicated in activating stem-like prostate tumor-initiating cells and activation of NFκB signaling, which is thought to contribute to the development of m-CRPC in AR-independent cells (24, 43). These studies support our findings that CXCL12γ induces a CSC phenotype through CXCR4-mediated activation of PKCα/NFκB signaling, which promotes prostate cancer tumor outgrowth and metastasis (44–47).

In addition, chemotherapy with docetaxel alters protein targets involved in cell survival, normal physiologic functions, and oncogenesis (48–50). Docetaxel also increases circulating cytokines in castration-resistant prostate cancer patients (49). CXCL12–CXCR4 signaling is known to prevent docetaxel-induced microtubule stabilization via p21-activated kinase 4 (PAK4)-dependent activation of LIM domain kinase 1 in prostate cancer cells (48). Activation of PKC signaling modulates microtubule cytoskeleton and cell morphology (39–41) and MARCKS as a downstream target of PKC signaling pathway is known to be critical for regulating multiple pathophysiologic processes, including chemoresistance (40–42). Recent evidence also shows that docetaxel reduces PKC activity by targeting of taxol-mediated suppression of NFκB (41, 42). These studies support our data demonstrating that intracellular CXCL12γ and CXCR4 interactions associate with the induction of a CSC phenotype and resistance to chemotherapy through PKCα/NFκB signaling pathways. Thus, understanding the biology of CSC expansion by therapy and by what mechanisms CSCs drive resistance to the cytotoxic effects will implicate the development of potential novel therapeutics.

Neuroendocrine prostate cancer disease has an extremely aggressive clinical course with a high mortality rate and is commonly observed in the late stages of the prostate cancer coincident with metastatic spread and resistance to treatment (10, 12–15, 35–38). Neuroendocrine cells may arise by two possible processes, the differentiation of CSCs into neuroendocrine cells, or transdifferentiation of preexisting adenocarcinoma cells. In the first instance, preexisting CSCs differentiate into cancer cells with three distinct phenotypes (luminal, basal, and neuroendocrine) or a cellular reprograming of differentiated epithelial cells to stem-like states poised for differentiating to progenitors. Cells with a neuroendocrine phenotype highly express the stem cell surface markers PROM1/CD133 and CD44 (10), which suggests that neuroendocrine cells share features with CSCs (14, 35). Recent studies show that the association of a neuroendocrine phenotype with CSC and EMT phenotypes such that when injected into nude mice, a highly vascularized tumor with a high density of neuroendocrine cells expressing low levels of E-cadherin and β-catenin and high levels of vimentin are formed (14, 35, 36).

As the second possible mechanism, the cells with a neuroendocrine phenotype arise from transdifferentiation of preexisting adenocarcinoma cells, which have accumulated multiple molecular alterations (14, 15, 51, 52). Growing evidence suggests that adenocarcinoma cells can undergo a transdifferentiation process to become neuroendocrine prostate cancer cells, which acquire a similar phenotype to normal neuroendocrine cells and express several neuroendocrine markers (53, 54). In this transdifferentiation process, cell-autonomous signaling pathways may facilitate acquisition of a neuroendocrine state, which in conjunction with microenvironmental clues promotes tumor outgrowth, survival, and therapeutic-resistant properties. Recent findings demonstrate that IL6, epinephrine, and forskolin induce neuroendocrine differentiation in prostate cancer cells through activation of intracellular cAMP and protein kinase A (PKA) suggests this possibility (55, 56). Together, these studies indicate that multiple signaling pathways are likely active in mechanisms associated with neuroendocrine differentiation. Furthermore, growing evidence also shows that deregulation of cellular signaling by genetic or epigenetic alterations are associated with neuroendocrine differentiation during therapies. Neuroendocrine prostate cancer is often associated with accumulation of new genetic alterations, such as loss of tumor suppressors (RB1, PTEN, and p53), TMPRSS2-ERG rearrangement, and expression of proto-oncogenes (AURKA, N-MYC, and BCL2; refs. 14, 29, 37, 38, 52, 57, 58). NDRG1 expression involves metastasis and neuroendocrine differentiation in prostate cancer cells by loss of RB in a hypoxia-dependent fashion, and these biologic events are associated with activation of the NFκB signaling pathway, which contributes to metastatic, castration-resistant, or neuroendocrine disease (58). In this investigation, our findings show that CXCL12γ induces N-MYC, NDRG1, and ENO2 expression in neuroendocrine phenotype of prostate cancer cells (37, 59), which contribute to aggressive disease.

Currently, there is a continued need to develop biomarkers that predict response to chemotherapy for m-CRPC patients. Although most studies have focused on the important role of AR during the transition to m-CRPC, these approaches do not explain the resistance process in conjunction with alternation of signaling pathways, suggesting that AR is not the only pathway involved in the formation of m-CRPC (7, 11). Moreover, CTC numbers have been shown to correlate with patient outcome with overall survival in m-CRPC. In fact, the AR splice variant 7 (AR-V7) is associated with resistance to both enzalutamide and abiraterone in prostate cancer cells and has been shown to be one potential marker for CTCs (4). Here, we show that CXCL12γ was significantly higher in men with increased ENO2 expression. Thus, CXCL12γ expression may serve as another potential marker of neuroendocrine prostate cancer disease and the development of m-CRPC.

Finally, our findings show for the first time that expression of the intracellular chemokine CXCL12γ facilitates the emergence of CSCs and neuroendocrine prostate cancer associated with metastatic progression, chemoresistance, and tumor recurrence. That the molecular signatures of CSCs have tumorigenic capability with a large proliferative potential suggests that CXCL12γ promotes a cellular reprograming of differentiated epithelial cancer cells to stem-like states poised for differentiation into alternative phenotypes.

No potential conflicts of interest were disclosed.

Conception and design: Y. Jung, F.C. Cackowski, J.K. Kim, K.J. Pienta, R.S. Taichman

Development of methodology: Y. Jung, F.C. Cackowski, A.M. Decker, Y. Wang

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Jung, F.C. Cackowski, K. Yumoto, A.M. Decker, J. Wang, E. Lee, Y. Wang, J.-S. Chung, A.M. Gursky, P.H. Krebsbach, T.M. Morgan, R.S. Taichman

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Jung, F.C. Cackowski, K. Yumoto, A.M. Decker, J. Wang, J.K. Kim, E. Lee, Y. Wang, J.-S. Chung, P.H. Krebsbach, T.M. Morgan, R.S. Taichman

Writing, review, and/or revision of the manuscript: Y. Jung, A.M. Decker, J.K. Kim, Y. Wang, P.H. Krebsbach, K.J. Pienta, T.M. Morgan, R.S. Taichman

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Jung, J. Wang, P.H. Krebsbach, R.S. Taichman

Study supervision: Y. Jung, P.H. Krebsbach, K.J. Pienta, R.S. Taichman

The authors wish to thank Taocong Jin and Sasha Meshinchi for technical and logistical support in Molecular Core and Microscopy & Image Analysis Laboratory (MIL), Comprehensive Cancer Center Microscopy Core at University of Michigan, Ann Arbor, MI. Dr. Juan C. Ramirez (National Centre for Cardiovascular Research, Spain) kindly provided pLV and pLV-CXCL12γ vectors and CXCL12γ antibody. This work is directly supported by the NCI (to R.S. Taichman; CA093900 and CA163124), the Department of Defense [to R.S. Taichman (W81XW-15-1-0413 and W81XWH-14-1-0403) and T.M. Morgan (W81XWH-14-1-0287)], and the Prostate Cancer Foundation Challenge Award (to R.S. Taichman; 16CHAL05). R.S. Taichman receives support as the Major McKinley Ash Collegiate Professor. F.C. Cackowski receives support from a Career Enhancement Award, Sub-Award (F048931) of NIH/NCI Prostate Cancer Specialized Program in Research Excellence (SPORE) to Arul Chinnaiyan at the University of Michigan (F036250).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Hotte
SJ
,
Saad
F
. 
Current management of castrate-resistant prostate cancer
.
Curr Oncol
2010
;
17
:
S72
9
.
2.
Pienta
KJ
,
Esper
PS
. 
Risk factors for prostate cancer
.
Ann Intern Med
1993
;
118
:
793
803
.
3.
Morgan
TM
,
Lange
PH
,
Porter
MP
,
Lin
DW
,
Ellis
WJ
,
Gallaher
IS
, et al
Disseminated tumor cells in prostate cancer patients after radical prostatectomy and without evidence of disease predicts biochemical recurrence
.
Clin Cancer Res
2009
;
15
:
677
83
.
4.
Qu
F
,
Xie
W
,
Nakabayashi
M
,
Zhang
H
,
Jeong
SH
,
Wang
X
, et al
Association of AR-V7 and prostate-specific antigen RNA levels in blood with efficacy of abiraterone acetate and enzalutamide treatment in men with prostate cancer
.
Clin Cancer Res
2017
;
23
:
726
34
.
5.
Scher
HI
,
Lu
D
,
Schreiber
NA
,
Louw
J
,
Graf
RP
,
Vargas
HA
, et al
Association of AR-V7 on circulating tumor cells as a treatment-specific biomarker with outcomes and survival in castration-resistant prostate cancer
.
JAMA Oncol
2016
;
2
:
1441
9
.
6.
Antonarakis
ES
,
Lu
C
,
Wang
H
,
Luber
B
,
Nakazawa
M
,
Roeser
JC
, et al
AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer
.
N Engl J Med
2014
;
371
:
1028
38
.
7.
Beltran
H
,
Beer
TM
,
Carducci
MA
,
de Bono
J
,
Gleave
M
,
Hussain
M
, et al
New therapies for castration-resistant prostate cancer: efficacy and safety
.
Eur Urol
2011
;
60
:
279
90
.
8.
Cojoc
M
,
Mäbert
K
,
Muders
MH
,
Dubrovska
A
. 
A role for cancer stem cells in therapy resistance: cellular and molecular mechanisms
.
Semin Cancer Biol
2015
;
31
:
16
27
.
9.
Dean
M
,
Fojo
T
,
Bates
S
. 
Tumour stem cells and drug resistance
.
Nat Rev Cancer
2005
;
5
:
275
84
.
10.
Borges
GT
,
Vencio
EF
,
Quek
SI
,
Chen
A
,
Salvanha
DM
,
Vencio
RZ
, et al
Conversion of prostate adenocarcinoma to small cell carcinoma-like by reprogramming
.
J Cell Physiol
2016
;
231
:
2040
7
.
11.
Ojo
D
,
Lin
X
,
Wong
N
,
Gu
Y
,
Tang
D
. 
Prostate cancer stem-like cells contribute to the development of castration-resistant prostate cancer
.
Cancers
2015
;
7
:
2290
308
.
12.
Chan
F
,
Goodman
O
,
Fink
L
,
Vogelzang
NJ
,
Pomerantz
D
,
Khoury
JD
. 
Dramatically elevated circulating tumor cell numbers in a patient with small cell neuroendocrine carcinoma of the prostate
.
Arch Pathol Lab Med
2010
;
134
:
120
3
.
13.
Nelson
E
,
Cambio
A
,
Yang
J
,
Ok
J
,
Lara
PN
,
Evans
CP
. 
Clinical implications of neuroendocrine differentiation in prostate cancer
.
Prostate Cancer Prostatic Dis
2007
;
10
:
6
14
.
14.
Conteduca
V
,
Aieta
M
,
Amadori
D
,
De Giorgi
U
. 
Neuroendocrine differentiation in prostate cancer: current and emerging therapy strategies
.
Crit Rev Oncol Hematol
2014
;
92
:
11
24
.
15.
Parimi
V
,
Goyal
R
,
Poropatich
K
,
Yang
XJ
. 
Neuroendocrine differentiation of prostate cancer: a review
.
Am J Clin Exp Urol
2014
;
2
:
273
.
16.
Kryczek
I
,
Wei
S
,
Keller
E
,
Liu
R
,
Zou
W
. 
Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis
.
Am J Physiol Cell Physiol
2007
;
292
:
C987
C95
.
17.
Begley
LA
,
MacDonald
JW
,
Day
ML
,
Macoska
JA
. 
CXCL12 activates a robust transcriptional response in human prostate epithelial cells
.
J Biol Chem
2007
;
282
:
26767
74
.
18.
Jung
Y
,
Kim
JK
,
Shiozawa
Y
,
Wang
J
,
Mishra
A
,
Joseph
J
, et al
Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis
.
Nat Commun
2013
;
4
:
1795
.
19.
Wang
Q
,
Diao
X
,
Sun
J
,
Chen
Z
. 
Stromal cell-derived factor-1 and vascular endothelial growth factor as biomarkers for lymph node metastasis and poor cancer-specific survival in prostate cancer patients after radical prostatectomy
.
Urol Oncol
2013
;
31
:
312
7
.
20.
Wang
J
,
Ying
G
,
Wang
J
,
Jung
Y
,
Lu
J
,
Zhu
J
, et al
Characterization of phosphoglycerate kinase-1 expression of stromal cells derived from tumor microenvironment in prostate cancer progression
.
Cancer Res
2010
;
70
:
471
80
.
21.
Wang
J
,
Loberg
R
,
Taichman
RS
. 
The pivotal role of CXCL12 (SDF-1)/CXCR4 axis in bone metastasis
.
Cancer Metastasis Rev
2006
;
25
:
573
87
.
22.
Sun
YX
,
Wang
J
,
Shelburne
CE
,
Lopatin
DE
,
Chinnaiyan
AM
,
Rubin
MA
, et al
Expression of CXCR4 and CXCL12 (SDF-1) in human prostate cancers (PCa) in vivo
.
J Cell Biochem
2003
;
89
:
462
73
.
23.
Sun
YX
,
Schneider
A
,
Jung
Y
,
Wang
J
,
Dai
J
,
Wang
J
, et al
Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo
.
J Bone Miner Res
2005
;
20
:
318
29
.
24.
Rajasekhar
VK
,
Studer
L
,
Gerald
W
,
Socci
ND
,
Scher
HI
. 
Tumour-initiating stem-like cells in human prostate cancer exhibit increased NF-kappaB signalling
.
Nat Commun
2011
;
2
:
162
.
25.
Cavnar
S
,
Ray
P
,
Moudgil
P
,
Chang
S
,
Luker
K
,
Linderman
J
, et al
Microfluidic source-sink model reveals effects of biophysically distinct CXCL12 isoforms in breast cancer chemotaxis
.
Integr Biol
2014
;
6
:
564
76
.
26.
Ray
P
,
Stacer
AC
,
Fenner
J
,
Cavnar
SP
,
Meguiar
K
,
Brown
M
, et al
CXCL12-gamma in primary tumors drives breast cancer metastasis
.
Oncogene
2015
;
34
:
2043
51
.
27.
Zhao
S
,
Chang
SL
,
Linderman
JJ
,
Feng
FY
,
Luker
GD
. 
A comprehensive analysis of CXCL12 isoforms in breast cancer1,2
.
Transl Oncol
2014
May 13.
[Epub ahead of print]. doi: 10.1016/j.tranon.2014.04.001
.
28.
Gahan
JC
,
Gosalbez
M
,
Yates
T
,
Young
EE
,
Escudero
DO
,
Chi
A
, et al
Chemokine and chemokine receptor expression in kidney tumors: molecular profiling of histological subtypes and association with metastasis
.
J Urol
2012
;
187
:
827
33
.
29.
Torres
R
,
Ramirez
JC
. 
A chemokine targets the nucleus: Cxcl12-gamma isoform localizes to the nucleolus in adult mouse heart
.
PLoS One
2009
;
4
:
e7570
.
30.
Yu
L
,
Cecil
J
,
Peng
SB
,
Schrementi
J
,
Kovacevic
S
,
Paul
D
, et al
Identification and expression of novel isoforms of human stromal cell-derived factor 1
.
Gene
2006
;
374
:
174
9
.
31.
Duhagon
MA
,
Hurt
EM
,
Sotelo-Silveira
JR
,
Zhang
X
,
Farrar
WL
. 
Genomic profiling of tumor initiating prostatospheres
.
BMC Genomics
2010
;
11
:
324
.
32.
Armstrong
AJ
,
Marengo
MS
,
Oltean
S
,
Kemeny
G
,
Bitting
R
,
Turnbull
J
, et al
Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers
.
Mol Cancer Res
2011
;
9
:
997
1007
.
33.
Reyes
EE
,
Gillard
M
,
Duggan
R
,
Wroblewski
K
,
Kregel
S
,
Isikbay
M
, et al
Molecular analysis of CD133-positive circulating tumor cells from patients with metastatic castration-resistant prostate cancer
.
J Transl Sci
2015
;
1
.
Available at
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4704802/.
34.
Barriere
G
,
Fici
P
,
Gallerani
G
,
Fabbri
F
,
Zoli
W
,
Rigaud
M
. 
Circulating tumor cells and epithelial, mesenchymal and stemness markers: characterization of cell subpopulations
.
Ann Transl Med
2014
;
2
:
109
.
35.
Palapattu
GS
,
Wu
C
,
Silvers
CR
,
Martin
HB
,
Williams
K
,
Salamone
L
, et al
Selective expression of CD44, a putative prostate cancer stem cell marker, in neuroendocrine tumor cells of human prostate cancer
.
Prostate
2009
;
69
:
787
98
.
36.
McKeithen
D
,
Graham
T
,
Chung
LW
,
Odero-Marah
V
. 
Snail transcription factor regulates neuroendocrine differentiation in LNCaP prostate cancer cells
.
Prostate
2010
;
70
:
982
92
.
37.
Lee
JK
,
Phillips
JW
,
Smith
BA
,
Park
JW
,
Stoyanova
T
,
McCaffrey
EF
, et al
N-Myc drives neuroendocrine prostate cancer initiated from human prostate epithelial cells
.
Cancer Cell
2016
;
29
:
536
47
.
38.
Beltran
H
,
Prandi
D
,
Mosquera
JM
,
Benelli
M
,
Puca
L
,
Cyrta
J
, et al
Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer
.
Nat Med
2016
;
22
:
298
305
.
39.
Goodnight
J
,
Mischak
H
,
Kolch
W
,
Mushinski
JF
. 
Immunocytochemical localization of eight protein kinase C isozymes overexpressed in NIH 3T3 fibroblasts. Isoform-specific association with microfilaments, Golgi, endoplasmic reticulum, and nuclear and cell membranes.
J Biol Chem
1995
;
270
:
9991
10001
.
40.
Yang
Y
,
Chen
Y
,
Saha
M
,
Chen
J
,
Evans
K
,
Qiu
L
, et al
Targeting phospho-MARCKS overcomes drug-resistance and induces antitumor activity in preclinical models of multiple myeloma
.
Leukemia
2015
;
29
:
715
26
.
41.
Chen
C-H
,
Cheng
C-T
,
Yuan
Y
,
Zhai
J
,
Arif
M
,
Fong
LWR
, et al
Elevated MARCKS phosphorylation contributes to unresponsiveness of breast cancer to paclitaxel treatment
.
Oncotarget
2015
;
6
:
15194
.
42.
Spencer
W
,
Kwon
H
,
Crepieux
P
,
Leclerc
N
,
Lin
R
,
Hiscott
J
. 
Taxol selectively blocks microtubule dependent NF-kB activation by phorbol ester via inhibition of IkBa phosphorylation and degradation
.
Oncogene
1999
;
18
:
495
506
.
43.
Trushin
SA
,
Pennington
KN
,
Carmona
EM
,
Asin
S
,
Savoy
DN
,
Billadeau
DD
, et al
Protein kinase Calpha (PKCalpha) acts upstream of PKCtheta to activate IkappaB kinase and NF-kappaB in T lymphocytes
.
Mol Cell Biol
2003
;
23
:
7068
81
.
44.
McCall
P
,
Bennett
L
,
Ahmad
I
,
Mackenzie
L
,
Forbes
I
,
Leung
H
, et al
NFκB signalling is upregulated in a subset of castrate-resistant prostate cancer patients and correlates with disease progression
.
Br J Cancer
2012
;
107
:
1554
63
.
45.
Jin
R
,
Yi
Y
,
Yull
FE
,
Blackwell
TS
,
Clark
PE
,
Koyama
T
, et al
NF-κB gene signature predicts prostate cancer progression
.
Cancer Res
2014
;
74
:
2763
72
.
46.
Jin
R
,
Yamashita
H
,
Yu
X
,
Wang
J
,
Franco
OE
,
Wang
Y
, et al
Inhibition of NF-kappa B signaling restores responsiveness of castrate-resistant prostate cancer cells to anti-androgen treatment by decreasing androgen receptor-variant expression
.
Oncogene
2015
;
34
:
3700
10
.
47.
Rehman
AO
,
Wang
CY
. 
CXCL12/SDF-1 alpha activates NF-kappaB and promotes oral cancer invasion through the Carma3/Bcl10/Malt1 complex
.
Int J Oral Sci
2009
;
1
:
105
18
.
48.
Bhardwaj
A
,
Srivastava
SK
,
Singh
S
,
Arora
S
,
Tyagi
N
,
Andrews
J
, et al
CXCL12/CXCR4 signaling counteracts docetaxel-induced microtubule stabilization via p21-activated kinase 4-dependent activation of LIM domain kinase 1
.
Oncotarget
2014
;
5
:
11490
500
.
49.
Mahon
K
,
Lin
H
,
Castillo
L
,
Lee
B
,
Lee-Ng
M
,
Chatfield
M
, et al
Cytokine profiling of docetaxel-resistant castration-resistant prostate cancer
.
Br J Cancer
2015
;
112
:
1340
8
.
50.
Li
Y
,
Li
X
,
Hussain
M
,
Sarkar
FH
. 
Regulation of microtubule, apoptosis, and cell cycle-related genes by taxotere in prostate cancer cells analyzed by microarray
.
Neoplasia
2004
;
6
:
158
67
.
51.
Zelivianski
S
,
Verni
M
,
Moore
C
,
Kondrikov
D
,
Taylor
R
,
Lin
M-F
. 
Multipathways for transdifferentiation of human prostate cancer cells into neuroendocrine-like phenotype
.
Biochim Biophys Acta
2001
;
1539
:
28
43
.
52.
Terry
S
,
Beltran
H
. 
The many faces of neuroendocrine differentiation in prostate cancer progression
.
Front Oncol
2014
;
4
:
60
.
53.
Yuan
T-C
,
Veeramani
S
,
Lin
M-F
. 
Neuroendocrine-like prostate cancer cells: neuroendocrine transdifferentiation of prostate adenocarcinoma cells
.
Endocr Relat Cancer
2007
;
14
:
531
47
.
54.
Hu
C-D
,
Choo
R
,
Huang
J
. 
Neuroendocrine differentiation in prostate cancer: a mechanism of radioresistance and treatment failure
.
Front Oncol
2015
;
14
:
90
.
55.
Qiu
Y
,
Robinson
D
,
Pretlow
TG
,
Kung
H-J
. 
Etk/Bmx, a tyrosine kinase with a pleckstrin-homology domain, is an effector of phosphatidylinositol 3′-kinase and is involved in interleukin 6-induced neuroendocrine differentiation of prostate cancer cells
.
Proc Natl Acad Sci U S A
1998
;
95
:
3644
9
.
56.
Bang
Y
,
Pirnia
F
,
Fang
W
,
Kang
W
,
Sartor
O
,
Whitesell
L
, et al
Terminal neuroendocrine differentiation of human prostate carcinoma cells in response to increased intracellular cyclic AMP
.
Proc Natl Acad Sci U S A
1994
;
91
:
5330
4
.
57.
Bandyopadhyay
S
,
Pai
SK
,
Hirota
S
,
Hosobe
S
,
Tsukada
T
,
Miura
K
, et al
PTEN up-regulates the tumor metastasis suppressor gene Drg-1 in prostate and breast cancer
.
Cancer Res
2004
;
64
:
7655
60
.
58.
Labrecque
MP
,
Takhar
MK
,
Nason
R
,
Santacruz
S
,
Tam
KJ
,
Massah
S
, et al
The retinoblastoma protein regulates hypoxia-inducible genetic programs, tumor cell invasiveness and neuroendocrine differentiation in prostate cancer cells
.
Oncotarget
2016
;
7
:
24284
.
59.
Lee
E
,
Wang
J
,
Yumoto
K
,
Jung
Y
,
Cackowski
FC
,
Decker
AM
, et al
DNMT1 regulates epithelial-mesenchymal transition and cancer stem cells, which promotes prostate cancer metastasis
.
Neoplasia
2016
;
18
:
553
66
.