Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Biomedicines ; 12(4)2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38672111

RESUMO

Interleukin-17 (IL-17) is a pro-inflammatory cytokine that participates in innate and adaptive immune responses and plays an important role in host defense, autoimmune diseases, tissue regeneration, metabolic regulation, and tumor progression. Post-translational modifications (PTMs) are crucial for protein function, stability, cellular localization, cellular transduction, and cell death. However, PTMs of IL-17 receptor A (IL-17RA) have not been investigated. Here, we show that human IL-17RA was targeted by F-box and WD repeat domain-containing 11 (FBXW11) for ubiquitination, followed by proteasome-mediated degradation. We used bioinformatics tools and biochemical techniques to determine that FBXW11 ubiquitinated IL-17RA through a lysine 27-linked polyubiquitin chain, targeting IL-17RA for proteasomal degradation. Domain 665-804 of IL-17RA was critical for interaction with FBXW11 and subsequent ubiquitination. Our study demonstrates that FBXW11 regulates IL-17 signaling pathways at the IL-17RA level.

2.
J Natl Cancer Inst ; 116(3): 421-433, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-37847647

RESUMO

BACKGROUND: Although the fusion of the transmembrane serine protease 2 gene (TMPRSS2) with the erythroblast transformation-specific-related gene (ERG), or TMPRSS2-ERG, occurs frequently in prostate cancer, its impact on clinical outcomes remains controversial. Roughly half of TMPRSS2-ERG fusions occur through intrachromosomal deletion of interstitial genes and the remainder via insertional chromosomal rearrangements. Because prostate cancers with deletion-derived TMPRSS2-ERG fusions are more aggressive than those with insertional fusions, we investigated the impact of interstitial gene loss on prostate cancer progression. METHODS: We conducted an unbiased analysis of transcriptome data from large collections of prostate cancer samples and employed diverse in vitro and in vivo models combined with genetic approaches to characterize the interstitial gene loss that imposes the most important impact on clinical outcome. RESULTS: This analysis identified FAM3B as the top-ranked interstitial gene whose loss is associated with a poor prognosis. The association between FAM3B loss and poor clinical outcome extended to fusion-negative prostate cancers where FAM3B downregulation occurred through epigenetic imprinting. Importantly, FAM3B loss drives disease progression in prostate cancer. FAM3B acts as an intermediator of a self-governing androgen receptor feedback loop. Specifically, androgen receptor upregulates FAM3B expression by binding to an intronic enhancer to induce an enhancer RNA and facilitate enhancer-promoter looping. FAM3B, in turn, attenuates androgen receptor signaling. CONCLUSION: Loss of FAM3B in prostate cancer, whether through the TMPRSS2-ERG translocation or epigenetic imprinting, causes an exit from this autoregulatory loop to unleash androgen receptor activity and prostate cancer progression. These findings establish FAM3B loss as a new driver of prostate cancer progression and support the utility of FAM3B loss as a biomarker to better define aggressive prostate cancer.


Assuntos
Neoplasias da Próstata , Receptores Androgênicos , Masculino , Humanos , Receptores Androgênicos/genética , Retroalimentação , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transcriptoma , Proteínas de Fusão Oncogênica/genética , Regulador Transcricional ERG/genética , Regulador Transcricional ERG/metabolismo , Proteínas de Neoplasias/genética , Citocinas/genética
3.
Lupus Sci Med ; 10(1)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37041033

RESUMO

OBJECTIVE: SLE is an autoimmune disease characterised by persistent inflammation and autoantibody production. Genetic predisposition and environmental factors such as a high-fat diet (HFD) may contribute to lupus development. However, the immune cell profile and gender difference in response to HFD in lupus have not been reported. Here we investigated the impact of HFD on lupus pathogenesis and autoimmunity using lupus-prone mice. METHODS: Thirty male and 30 female MRL/lymphoproliferation (lpr) mice were fed with regular diet (RD) or HFD. Body weights were recorded weekly. SLE progression was monitored by skin lesion, urine protein, titres of antidouble-strand DNA (dsDNA) and ANA. At week 14, kidney and skin tissue sections were stained with H&E and periodic acid-Schiff to detect histological kidney index and skin score. Splenocytes were identified by immunofluorescence staining and flow cytometry. RESULTS: HFD significantly increased body weight and lipid levels compared with RD (p<0.01). Skin lesions were observed in 55.6% of the HFD group compared with 11.1% of the RD group, with greater histopathological skin scores in the female HFD group (p<0.01). Although both male and female mice had higher serum IgG in the HFD group than in the RD group, only the male HFD group showed an increased trend in anti-dsDNA Ab and ANA titres. Kidney pathological changes in the HFD group were more severe in male mice than in female mice (p<0.05), detected by proteinuria, kidney index and glomerular cell proliferation. Significant increases of germinal centre B cells and T follicular helper cells were observed in the spleens of HFD mice (p<0.05). CONCLUSION: HFD induced an accelerated and exacerbated lupus development and autoimmunity in MRL/lpr mice. Our results parallel many known clinical lupus phenotypes and sexual dimorphism in which male patients are likelier to have a severe disease (nephritis) than female lupus patients who may have a broader range of lupus symptoms.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Masculino , Feminino , Humanos , Animais , Camundongos , Autoimunidade , Dieta Hiperlipídica , Camundongos Endogâmicos MRL lpr , Obesidade
4.
Biomedicines ; 10(8)2022 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-36009523

RESUMO

Patients with psoriasis tend to develop skin cancer, and the hyperproliferation of the epidermis is a histopathological hallmark of both psoriasis and cutaneous squamous cell carcinoma (SCC), indicating that they may share pathogenic mechanisms. Interleukin-17 (IL17) stimulates the proliferation of the epidermis, leading to psoriasis. Overexpression of Polo-like kinase 4 (PLK4), which controls centriole duplication, has been identified in SCC, which also shows the hyperproliferation of keratinocytes. To investigate the cooperation between IL17 signaling and centriole duplication in epidermal proliferation, we established psoriasis and skin papilloma models in wild type (WT), IL17 receptor A (T779A) knockin (Il17ra(T779A)-KI), and IL17 receptor C knockout (Il17rc-KO) mouse strains. Bioinformatics, Western blot, immunohistochemical staining, colony formation, and real-time PCR were used to determine the effect of IL17 signaling and centrinone on epithelial proliferation. In the psoriasis model, compared to WT and Il17ra(T779A)-KI, Il17rc-KO dramatically suppressed epidermal thickening. The proliferation of keratinocytes significantly decreased in this order from WT to Il17ra(T779A)-KI and Il17rc-KO mice. In the skin papilloma model, Il17ra(T779A)-KI significantly decreased tumor burden compared to the WT, while Il17rc-KO abolished papilloma development. However, centrinone, a selective inhibitor of PLK4, did not affect skin lesion formation in either model. Our data demonstrated that Il17ra(T779A)-KI and Il17rc-KO prevent the development of psoriasis and tumorigenesis in the skin, while the topical administration of centrinone does not have any effect.

5.
Exp Cell Res ; 417(1): 113210, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35597298

RESUMO

Cancer cells acquire immunoediting ability to evade immune surveillance and thus escape eradication. It is widely known that mutant proteins encoded from tumor suppressor TP53 exhibit gain-of-function in cancer cells, thereby promoting progression; however, how mutant p53 contributes to the sheltering of cancer cells from host anticancer immunity remains unclear. Herein, we report that murine p53 missense mutation G242A (corresponding to human G245A) suppresses the activation of host natural killer (NK) cells, thereby enabling breast cancer cells to avoid immune assault. We found that serial injection of EMT6 breast cancer cells that carry wild-type (wt) Trp53, like normal fibroblasts, promoted NK activity in mice, while SVTneg2 cells carrying Trp53 G242A+/+ mutation decreased NK cell numbers and increased CD8+ T lymphocyte numbers in spleen. Innate immunity based on NK cells and CD8 T cells was reduced in p53 mutant-carrying transgenic mice (Trp53 R172H/+, corresponding to human R175H/+). Further, upon co-culture with isolated NK cells, EMT6 cells substantively activated NK cells and proliferation thereof, increasing interferon-gamma (IFN-γ) production; however, SVTneg2 cells suppressed NK cell activation. Further mechanistic study elucidated that p53 can modulate expression by cancer cells of Mult-1 and H60a, which are activating and inhibitory ligands for NKG2D receptors of NK cells, respectively, to enhance immune surveillance against cancer. Our findings demonstrate that wt p53 is requisite for NK cell-based immune recognition and elimination of cancerous cells, and perhaps more importantly, that p53 missense mutant presence in cancer cells impairs NK cell-attributable responses, thus veiling cancerous cells from host immunity and enabling cancer progression.


Assuntos
Neoplasias da Mama , Células Matadoras Naturais , Proteína Supressora de Tumor p53 , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Feminino , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Transgênicos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Biochem Biophys Rep ; 29: 101200, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35036586

RESUMO

Growth differentiation factor 5 (Gdf5) and doublecortin (Dcx) genes are both expressed in joint interzone cells during synovial joint development. In this study, we re-analyzed the single cell RNA-sequencing data (Gene Expression Omnibus GSE151985) generated from Gdf5 + cells of mouse knee joints at embryonic stages of E12.5, E13.5, E14.5, and E15.5, with a new focus on Dcx. We found that Dcx expression was enriched in clusters of Gdf5 + cells, with high expression levels of pro-chondrogenic genes including sex determining region Y-box transcription factor 5 (Sox5), Sox6, Sox9, Gdf5, versican, matrilin 4, collagen type II α 1 chain (Col2a1), Col9a1, Col9a2, and Col9a3 at E12.5. Dcx + and Dcx - cells had differential gene expression profiles. The up-regulated genes in Dcx + vs. Dcx - cells at E12.5 and E13.5 were enriched in chondrocyte differentiation and cartilage development, whereas those genes up-regulated at E14.5 and E15.5 were enriched in RNA splicing, protein stability, cell proliferation, and cell growth. Gene expression profiles in Dcx + cells showed rapid daily changes from E12.5 to E15.5, with limited number of genes shared across the time period. Expression of Gdf5, Sox5, Sox6, melanoma inhibitory activity, noggin, odd-skipped related transcription factor 2, matrilin 4, and versican was positively correlated with Dcx expression. Our results demonstrate that Dcx expression defines a subpopulation of Gdf5 + cells with chondrogenic potentials in E12.5 mouse embryonic limbs.

7.
NPJ Regen Med ; 6(1): 82, 2021 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-34848747

RESUMO

Localized cartilage lesions in early osteoarthritis and acute joint injuries are usually treated surgically to restore function and relieve pain. However, a persistent clinical challenge remains in how to repair the cartilage lesions. We expressed doublecortin (DCX) in human adipose-derived stromal/stem cells (hASCs) and engineered hASCs into cartilage tissues using an in vitro 96-well pellet culture system. The cartilage tissue constructs with and without DCX expression were implanted in the knee cartilage defects of rabbits (n = 42) and monkeys (n = 12). Cohorts of animals were euthanized at 6, 12, and 24 months after surgery to evaluate the cartilage repair outcomes. We found that DCX expression in hASCs increased expression of growth differentiation factor 5 (GDF5) and matrilin 2 in the engineered cartilage tissues. The cartilage tissues with DCX expression significantly enhanced cartilage repair as assessed macroscopically and histologically at 6, 12, and 24 months after implantation in the rabbits and 24 months after implantation in the monkeys, compared to the cartilage tissues without DCX expression. These findings suggest that hASCs expressing DCX may be engineered into cartilage tissues that can be used to treat localized cartilage lesions.

9.
Am J Clin Exp Urol ; 8(4): 93-100, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929404

RESUMO

Human prostate cancer often metastasizes to the bone, but the mechanisms are not quite clear. The difficulties in studying the biology of bone metastasis are due to lack of animal models with high frequency of bone metastases. In the present study, we tested two intra-arterial injection methods, i.e., intra-caudal artery injection and intra-femoral artery injection. Mouse prostate cancer cell line MPC3-luc was injected into C57BL/6J male mice via intra-caudal artery injection (n = 8) or intra-femoral artery injection (n = 11). We found one mouse developed metastatic tumors in both hind limbs and the tail after intra-caudal artery injection. Two mice developed metastatic tumors in the hind limb after intra-femoral artery injection. The metastatic tumors were detected by bioluminescent imaging and X-ray, and confirmed by histological examination. Our study finds that intra-arterial (either caudal or femoral artery) injection may be a useful model in studying prostate cancer bone metastasis, however, the injection technique is difficult.

10.
Am J Clin Exp Urol ; 8(4): 116-128, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929407

RESUMO

F-box and WD repeat domain containing (FBXW) family of E3 ligases has 10 members that ubiquitinate substrate proteins for proteasome-mediated degradation. Publicly archived datasets from The Cancer Genome Atlas (TCGA), Prostate Cancer Transcriptome Atlas (PCTA), and cBioPortal were analyzed for mRNA expression and genetic alterations of 10 FBXW genes. We found that FBXW7 mRNA expression was significantly decreased in primary prostate cancers compared to normal prostate tissues, whereas mRNA expression of FBXW8-10 was significantly increased in primary prostate cancers compared to normal prostate tissues. FBXW7 mRNA expression was also significantly decreased in breast invasive carcinoma, glioblastoma multiforme, head and neck squamous cell carcinoma, lung squamous cell carcinoma, and uterine corpus endometrial carcinoma. In contrast, FBXW7 mRNA expression was significantly increased in cholangiocarcinoma, colon adenocarcinoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, pheochromocytoma and paraganglioma, and thyroid carcinoma. Compared to normal tissues, FBXW5 mRNA expression was significantly increased in breast invasive carcinoma, cholangiocarcinoma, kidney chromophobe, kidney renal clear cell carcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, prostate adenocarcinoma, thyroid carcinoma, and uterine corpus endometrial carcinoma, whereas FBXW5 mRNA expression was only significantly decreased in colon adenocarcinoma. There were not any significant differences in gene copy number gains, losses, or gene simple somatic mutations between primary prostate cancers and normal prostate tissues. The mRNA expression levels of FBXW5, 7, 8, 9, and 12 were significantly higher in metastatic castration-resistant prostate cancers (mCRPCs) than primary prostate cancers, whereas mRNA expression levels of FBXW1 and 4 were significantly lower in mCRPCs than primary prostate cancers. All 10 FBXW genes had significantly more overall gene alterations including gene amplifications in mCRPCs than primary prostate cancers. FBXW5 and 7 had significantly more gene deep deletions in mCRPCs than primary prostate cancers and FBXW7 had significantly more gene missense mutations in mCRPCs than primary prostate cancers. Our findings suggest that different FBXW genes have differential mRNA expression in prostate cancer and other cancer types and their gene amplifications are significantly more in mCRPCs than primary prostate cancers. FBXW7 mRNA expression is consistently decreased in primary prostate cancers compared to normal prostate tissues.

11.
Am J Clin Exp Urol ; 7(4): 249-261, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31511831

RESUMO

Interleukin-17 (IL-17) has been demonstrated to promote development of a variety of cancers including prostate cancer in genetically modified mouse models. IL-17 is the main product secreted by T helper 17 (Th17) cells. A recent study has shown that Th17 cells and related genes are upregulated in human prostate cancers. However, there is no direct experimental evidence to demonstrate Th17's role in prostate cancer. In the present study, we co-implanted mouse prostate cancer MPC3-luc cells with Th17-polarized mouse splenocytes in the prostate of immunocompetent C57BL/6J male mice. We found that Th17-polarized splenocytes promoted orthotopic allograft prostate tumor growth compared to the control splenocytes. The numbers of IL-17-positive lymphocytes and macrophages were higher in the prostate tumors grown from co-implantation of MPC3-luc cells and Th17-polarized splenocytes, compared to the prostate tumors grown from co-implantation of MPC3-luc cells and control splenocytes. Our findings provide the first direct experimental evidence that Th17 cells may promote prostate cancer growth.

12.
Front Oncol ; 9: 546, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281798

RESUMO

Interleukin-17 (IL-17) has been shown to promote development of prostate, colon, skin, lung, breast, and pancreatic cancer. The purpose of this study was to determine if IL-17 regulates MTA1 expression and its biological consequences. Human cervical cancer HeLa and human prostate cancer DU-145 cell lines were used to test if IL-17 regulates metastasis associated 1 (MTA1) mRNA and protein expression using quantitative reverse transcription-polymerase chain reaction and Western blot analysis, respectively. Cell migration and invasion were studied using wound healing assays and invasion chamber assays. Thirty-four human cervical tissues were stained for IL-17 and MTA1 using immunohistochemical staining. We found that IL-17 increased MTA1 mRNA and protein expression in both cell lines. Cell migration was accelerated by IL-17, which was abolished by knockdown of MTA1 expression with small interference RNA (siRNA). Further, cell invasion was enhanced by IL-17, which was eliminated by MTA1 knockdown. Human cervical intra-epithelial neoplasia (CIN) and cervical cancer tissues had increased number of IL-17-positive cells and MTA1 expression compared to normal cervical tissues. The number of IL-17-positive cells was positively correlated with MTA1 expression. These findings demonstrate that IL-17 upregulates MTA1 mRNA and protein expression to promote HeLa and DU-145 cell migration and invasion.

13.
Am J Clin Exp Urol ; 7(3): 159-169, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31317055

RESUMO

Immunotherapy targeting programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) has shown efficacy in a variety of solid tumors. However, prostate cancer has often been a non-responder to anti-PD-1/PD-L1 therapies. The objective of this study was to determine PD-1 and PD-L1 expression status and its correlation with clinical features of the patients. A total of 279 patients who underwent radical prostatectomy for prostate cancer were included in this study. PD-1 and PD-L1 expression in primary prostate tumors was detected using immunohistochemical staining. Analyses were made between PD-1/PD-L1 status and patients' age, ethnicity, body mass index (BMI), diabetes mellitus, tumor stage, lymph node metastasis, prostate-specific antigen (PSA), Gleason score, grade group, and survival. We found that 6.5 (standard deviation 14.3; range 0-161.6) tumor-infiltrating lymphocytes per high power field were positive for PD-1 staining and 50/279 (17.9%) tumors were positive for PD-L1 staining. PD-L1-positive tumors had significantly more PD-1-positive lymphocytes than PD-L1-negative tumors. The number of PD-1-positive lymphocytes was not correlated with any clinical features except that patients with diabetes had significantly less PD-1-positive lymphocytes than patients without diabetes. In contrast, more PD-L1-positive tumors were found in older patients (≥ 65 years), obese patients (BMI ≥ 30), and patients with advanced tumor stage, lymph node metastasis, and high Gleason score. Neither PD-1 nor PD-L1 status was correlated with ethnicity, PSA, or survival. Our findings suggest that PD-L1 instead of PD-1 status is associated with the clinical features in human primary prostate tumors.

14.
Am J Clin Exp Urol ; 7(6): 352-377, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31970232

RESUMO

Interleukin-17 (IL-17) has been shown to promote development of hormone-naïve prostate cancer (HNPC) and castration-resistant prostate cancer (CRPC) as well as lymph node metastasis in mouse models. Gene alterations of IL-17 family of cytokines and their downstream genes in human prostate cancer have not been investigated. We studied 7 datasets archived in cBioPortal and queried gene alterations in a total of 1303 cases of human prostate cancers. 35 genes were examined, including IL-17 family of cytokines and receptors, IL-17-downstream genes, and genes related to IL-17-downstream genes. We found that 34/35 (97%) genes had significantly more alterations in metastatic prostate cancer (with alteration rates ranging from 3.42% to 13.01%) than primary prostate cancer (with alteration rates ranging from 0.40% to 2.96%). 15/35 (43%) genes had significantly more alterations in primary CRPC than primary HNPC. 34/35 (97%) genes had significantly more alterations in metastatic CRPC than primary HNPC. Only three genes (S100A7, S100A8, and S100A9) had significantly more alterations in metastatic CRPC than primary CRPC. The gene alterations were mostly gene amplifications (97%), while gene deep deletions, missense mutations, and truncating mutations were very rare. 7/35 (20%) genes had significantly more alterations in primary neuroendocrine prostate cancer (NEPC) than primary adenocarcinoma (AC). 23/35 (66%) genes had significantly more alterations in metastatic NEPC than metastatic AC. Only three genes (S100A7, S100A8, and S100A9) had significantly more alterations in metastatic NEPC than metastatic AC with neuroendocrine features. Most of the gene alterations in metastatic NEPC were gene amplifications (80%), while gene deep deletions, missense mutations, and truncating mutations were very rare. Our findings suggest that gene amplifications of IL-17 and related genes are more frequently found in metastatic CRPC and NEPC than primary hormone-naïve prostate adenocarcinomas, implying that IL-17 and related genes may play important roles in the progression from HNPC to CRPC and from primary location to metastasis as well as in development of metastatic NEPC.

15.
Am J Clin Exp Urol ; 6(3): 114-122, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30038943

RESUMO

Metastasis of prostate cancer causes substantial morbidity and mortality. The role of chronic inflammatory factors in promoting the development of prostate cancer metastasis remains unexamined due to a lack of immunocompetent animal models. Here we report an orthotopic mouse allograft model of prostate cancer that was used to assess interleukin-17's role in prostate cancer metastasis. A luciferase gene was stably introduced into a mouse prostate cancer cell line MPC3, named as MPC3-luc. MPC3-luc cells were mixed with Matrigel™ and inoculated into C57BL/6 mouse prostates, with recombinant mouse interleukin-17 (IL-17) (treatment group) or without IL-17 (control group). Bioluminescent imaging was used to track the growth and metastasis of prostate cancer metastasis. Immunohistochemistry was performed to confirm metastasis. Mice in the IL-17 treatment group had significantly higher incidence of metastasis than mice in the control group. However, there was no detectable difference in primary prostate tumor growth. Metastases were confirmed as originating from prostate cancer through staining for luciferase protein expression. Our findings suggest that interleukin-17 promotes prostate cancer metastasis in an orthotopic mouse allograft model.

16.
Am J Clin Exp Urol ; 5(3): 25-33, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29181435

RESUMO

Organoids mimic the architecture and functions of a small organ. Organoid culture technique has been rapidly accepted by all research communities during the past decade to study stem cells, organ development and function, and patient-specific diseases. A protocol for organoid culture of human and mouse prostate epithelial and cancer tissues has been reported. However, organoid culture of the commonly used human prostate cancer cell lines has yet to be established. We followed the published protocol and performed organoid culture of LNCaP and C4-2B cells in MatrigelTM and organoid culture medium for 14 days. We found that both LNCaP and C4-2B cell lines formed organoids that presented glandular structures. The cells within the organoids were androgen receptor-positive adenocarcinoma cells, but not p63-positive basal cells. The cells in the organoids responded to interleukin-17A treatment differently from the cells in the monolayer culture. The present study suggests that LNCaP and C4-2B cells are able to form organoids under the defined organoid culture conditions.

17.
Am J Clin Exp Urol ; 5(1): 1-9, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28337464

RESUMO

Approximately 36.5% of the U.S. adults (≥ 20 years old) are obese. Obesity has been associated with type 2 diabetes mellitus, cardiovascular disease, stroke, and several types of cancer. The present study included 1788 prostate cancer patients who were treated with radical prostatectomy at the Ochsner Health System, New Orleans, Louisiana, from January, 2001 to March, 2016. The patient's medical records were retrospectively reviewed. Body mass index (BMI), age, ethnicity (Caucasians versus African Americans), clinical stage, Gleason score, and prostate-specific antigen (PSA) levels were retrieved. The relative risk of the patients was stratified into low risk and high risk groups. Associative analyses found that BMI was associated with age, clinical stage, Gleason score, but not ethnicity, PSA levels, or the relative risk in this cohort. Age was associated with ethnicity, clinical stage, Gleason score, and PSA levels, as well as the relative risk. Ethnicity was associated with Gleason score and PSA levels as well as the relative risk, but not clinical stage. These findings suggest that obesity is associated with advanced prostate cancer with stage T3 or Gleason score ≥ 7 diseases, and age and ethnicity are important factors that are associated with the clinical features of prostate cancer patients.

18.
Immunol Lett ; 184: 7-14, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28223102

RESUMO

Programmed cell death protein 1 (PD-1) acts on PD-1 ligands (PD-L1 and PD-L2) to suppress activation of cytotoxic T lymphocytes. Interleukin-17 (IL-17) and tumor necrosis factor-α (TNF-α) are co-expressed by T helper 17 (TH17) cells in many tumors. The purpose of this study was to test if IL-17 and TNF-α may synergistically induce PD-L1 expression in human prostate cancer LNCaP and human colon cancer HCT116 cell lines. We found that IL-17 did not induce PD-L1 mRNA expression, but up-regulated PD-L1 protein expression in HCT116 and LNCaP cells. TNF-α induced PD-L1 mRNA and protein expression in both cell lines. Neither IL-17 nor TNF-α induced PD-L2 mRNA or protein expression. IL-17 and TNF-α acted individually rather than cooperatively in induction of PD-L1 expression. IL-17 and/or TNF-α activated AKT, nuclear factor-κB (NF-κB), and extracellular signal-regulated kinases 1/2 (ERK1/2) signaling pathways in HCT116 cells, whereas only NF-κB signaling was activated in LNCaP cells. NF-κB inhibitor could diminish PD-L1 protein expression induced by IL-17 and/or TNF-α in both HCT116 and LNCaP cell lines. ERK1/2 inhibitor could also reduce PD-L1 protein expression induced by IL-17 and/or TNF-α in HCT116 cells, while AKT inhibitor could abolish PD-L1 protein expression induced by IL-17 and/or TNF-α in LNCaP cells. These results suggest that IL-17 and TNF-α act individually rather than cooperatively through activation of NF-κB and ERK1/2 signaling to up-regulate PD-L1 expression in HCT116 cells, while the two inflammatory cytokines act through activation of NF-κB signaling, in the presence of AKT activity, to up-regulate PD-L1 expression in LNCaP cells.


Assuntos
Antígeno B7-H1/genética , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Interleucina-17/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Citocinas/metabolismo , Citocinas/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/farmacologia , Interleucina-17/farmacologia , Masculino , NF-kappa B/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
19.
Prostate ; 77(8): 888-899, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28240383

RESUMO

BACKGROUND: Chronic inflammation has been associated with the development and progression of human cancers including prostate cancer. The exact role of the inflammatory Th17-IL-17 pathway in prostate cancer remains unknown. In this study, we aimed to determine the importance of Th17 cells and IL-17 in a Pten-null prostate cancer mouse model. METHODS: The Pten-null mice were treated by Th17 inhibitor SR1001 or anti-mouse IL-17 monoclonal antibody from 6 weeks of age up to 12 weeks of age. For SR1001 treatment, the mice were injected intraperitoneally (i.p.) twice a day with vehicle or SR1001, which was dissolved in a dimethylsulfoxide (DMSO) solution. All mice were euthanized for necropsy at 12 weeks of age. For IL-17 antibody treatment, the mice were injected intravenously (i.v.) once every two weeks with control IgG or rat anti-mouse IL-17 monoclonal antibody, which was dissolved in PBS. The injection time points were at 6, 8, and 10 weeks old. All mice were analyzed for the prostate phenotypes at 12 weeks of age. RESULTS: We found that either SR1001 or anti-IL-17 antibody treatment decreased the formation of micro-invasive prostate cancer in Pten-null mice. The SR1001 or anti-IL-17 antibody treated mouse prostates had reduced proliferation, increased apoptosis, and reduced angiogenesis, as well as reduced inflammatory cell infiltration. By assessing the epithelial-to-mesenchymal transition (EMT) markers, we found that SR1001 or anti-IL-17 antibody treated prostate tissues had weaker EMT phenotype compared to the control treated prostates. CONCLUSIONS: These results demonstrated that Th17-IL-17 pathway plays a key role in prostate cancer progression in Pten-null mice. Targeting Th17-IL-17 pathway could prevent micro-invasive prostate cancer formation in mice. Prostate 77:888-899, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Anticorpos Monoclonais/farmacologia , Inflamação/imunologia , Interleucina-17 , Invasividade Neoplásica , Neoplasias da Próstata , Sulfonamidas/farmacologia , Células Th17/imunologia , Tiazóis/farmacologia , Animais , Modelos Animais de Doenças , Monitoramento de Medicamentos/métodos , Fatores Imunológicos/farmacologia , Injeções Intraperitoneais , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Masculino , Camundongos , Terapia de Alvo Molecular/métodos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Próstata/imunologia , Próstata/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Resultado do Tratamento
20.
Int J Gynecol Cancer ; 27(2): 196-205, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27870715

RESUMO

OBJECTIVE: Estrogen is a well-known oncogenic driver in endometrial (ECs) and breast cancers (BCs). Programmed cell death protein 1 (PD-1) and its ligands PD-1 Ligand 1 (PD-L1) and PD-L2 have been shown to mediate immune evasion of the tumor cells. The purpose of the present study was to assess the effects of estrogen on PD-L1 and PD-L2 expression in EC and BC cell lines. METHODS: 17ß-Estradiol (E2)-induced expression of PD-L1 and PD-L2 and possible signaling pathway were investigated in EC and BC cells. Coculture of T cells and cancer cells with E2 stimulation was performed to assess the functions of T cells. RESULTS: We found that E2 increased expression of PD-L1, but not PD-L2, protein via activation of phosphoinositide 3-kinase (PI3K)/Akt pathway in Ishikawa and Michigan Cancer Foundation-7 (MCF-7) cells. Phosphoinositide 3-kinase and Akt inhibitors could block E2's effects. 17ß-Estradiol did not increase PD-L1 mRNA transcription, but stabilized PD-L1 mRNA. 17ß-Estradiol's effects were only observed in estrogen receptor α (ERα)-positive Ishikawa and MCF-7 cells, but not in ERα-negative MDA-MB-231 cells. Coculture of Ishikawa or MCF-7 cells with T cells inhibited expression of interferon-γ and interleukin-2 and increased BCL-2-interacting mediator of cell death expression in the presence of E2. CONCLUSIONS: This study provides the first evidence that estrogen upregulates PD-L1 protein expression in ERα-positive EC and BC cells to suppress immune functions of T cells in the tumor microenvironment, demonstrating a new mechanism of how estrogen drives cancer progression.


Assuntos
Antígeno B7-H1/biossíntese , Neoplasias da Mama/metabolismo , Neoplasias do Endométrio/metabolismo , Estradiol/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Antígeno B7-H1/genética , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/imunologia , Receptor alfa de Estrogênio/biossíntese , Feminino , Humanos , Células Jurkat , Células MCF-7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA