Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Pathol ; 260(2): 177-189, 2023 06.
Article in English | MEDLINE | ID: mdl-36825524

ABSTRACT

Benign prostatic hyperplasia (BPH) occurs progressively with aging in men and drives deteriorating symptoms collectively known as lower urinary tract symptoms (LUTS). Age-associated changes in circulating steroid hormones, and prostate inflammation have been postulated in the etiology of BPH/LUTS. The link between hormones and inflammation in the development of BPH/LUTS is conflicting because they may occur independently or as sequential steps in disease pathogenesis. This study aimed to decipher the prostatic immune landscape in a mouse model of lower urinary tract dysfunction (LUTD). Steroid hormone imbalance was generated by the surgical implantation of testosterone (T) and estradiol (E2) pellets into male C57BL/6J mice and gene expression analysis was performed on ventral prostates (VPs). These experiments identified an increase in the expression of macrophage markers and Spp1/osteopontin (OPN). Localization studies of OPN pinpointed that OPN+ macrophages travel to the prostate lumen and transition into lipid-accumulating foam cells. We also observed a significant increase in the number of tissue macrophages in the VP which was prevented in OPN-knockout (OPN-KO) mice. In contrast, mast cells, but not macrophages, were significantly elevated in the dorsal prostate of T + E2-treated mice which was diminished in OPN-KO mice. Steroid hormone implantation progressively increased urinary frequency, which was ameliorated in OPN-KO mice. Our study underscores the role of age-associated steroid hormone imbalances as a mechanism of expanding the prostatic macrophage population, their luminal translocation, and foam cell differentiation. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Prostate , Prostatic Hyperplasia , Humans , Male , Mice , Animals , Prostate/pathology , Prostatic Hyperplasia/pathology , Osteopontin/genetics , Osteopontin/metabolism , Mice, Inbred C57BL , Testosterone , Inflammation , Cell Differentiation
2.
Proc Natl Acad Sci U S A ; 115(47): 12028-12033, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30373845

ABSTRACT

The effects of the growth hormone-releasing hormone (GHRH) agonist MR409 on various human cancer cells were investigated. In H446 small cell lung cancer (SCLC) and HCC827 and H460 (non-SCLC) cells, MR409 promoted cell viability, reduced cell apoptosis, and induced the production of cellular cAMP in vitro. Western blot analyses showed that treatment of cancer cells with MR409 up-regulated the expression of cyclins D1 and D2 and cyclin-dependent kinases 4 and 6, down-regulated p27kip1, and significantly increased the expression of the pituitary-type GHRH receptor (pGHRH-R) and its splice-variant (SV1). Hence, in vitro MR409 exerts agonistic action on lung cancer cells in contrast to GHRH antagonists. However, in vivo, MR409 inhibited growth of lung cancers xenografted into nude mice. MR409 given s.c. at 5 µg/day for 4 to 8 weeks significantly suppressed growth of HCC827, H460, and H446 tumors by 48.2%, 48.7%, and 65.6%, respectively. This inhibition of tumor growth by MR409 was accompanied by the down-regulation of the expression of pGHRH-R and SV1 in the pituitary gland and tumors. Tumor inhibitory effects of MR409 in vivo were also observed in other human cancers, including gastric, pancreatic, urothelial, prostatic, mammary, and colorectal. This inhibition of tumor growth parallel to the down-regulation of GHRH-Rs is similar and comparable to the suppression of sex hormone-dependent cancers after the down-regulation of receptors for luteinizing hormone-releasing hormone (LHRH) by LHRH agonists. Further oncological investigations with GHRH agonists are needed to elucidate the underlying mechanisms.


Subject(s)
Receptors, Neuropeptide/drug effects , Receptors, Pituitary Hormone-Regulating Hormone/drug effects , Sermorelin/analogs & derivatives , Alternative Splicing/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Female , Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/pharmacology , Humans , Mice , Mice, Nude , RNA Splicing/drug effects , Sermorelin/metabolism , Sermorelin/pharmacology , Small Cell Lung Carcinoma/metabolism , Xenograft Model Antitumor Assays/methods
3.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830342

ABSTRACT

Fibrogenic and inflammatory processes in the prostate are linked to the development of lower urinary tract symptoms (LUTS) in men. Our previous studies identified that osteopontin (OPN), a pro-fibrotic cytokine, is abundant in the prostate of men with LUTS, and its secretion is stimulated by inflammatory cytokines potentially to drive fibrosis. This study investigates whether the lack of OPN ameliorates inflammation and fibrosis in the mouse prostate. We instilled uropathogenic E. coli (UTI89) or saline (control) transurethrally to C57BL/6J (WT) or Spp1tm1Blh/J (OPN-KO) mice and collected the prostates one or 8 weeks later. We found that OPN mRNA and protein expression were significantly induced by E. coli-instillation in the dorsal prostate (DP) after one week in WT mice. Deficiency in OPN expression led to decreased inflammation and fibrosis and the prevention of urinary dysfunction after 8 weeks. RNAseq analysis identified that E. coli-instilled WT mice expressed increased levels of inflammatory and fibrotic marker RNAs compared to OPN-KO mice including Col3a1, Dpt, Lum and Mmp3 which were confirmed by RNAscope. Our results indicate that OPN is induced by inflammation and prolongs the inflammatory state; genetic blockade of OPN accelerates recovery after inflammation, including a resolution of prostate fibrosis.


Subject(s)
Escherichia coli Infections/genetics , Osteopontin/genetics , Prostate/metabolism , Urinary Tract Infections/genetics , Uropathogenic Escherichia coli/pathogenicity , Animals , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Disease Models, Animal , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Escherichia coli Infections/prevention & control , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibrosis , Gene Expression Regulation , Humans , Inflammation , Lumican/genetics , Lumican/metabolism , Male , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/deficiency , Prostate/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Urinary Tract Infections/metabolism , Urinary Tract Infections/pathology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/growth & development
4.
Prostate ; 80(13): 1058-1070, 2020 09.
Article in English | MEDLINE | ID: mdl-32692871

ABSTRACT

BACKGROUND: Most prostate cancers express androgen receptor (AR), and our previous studies have focused on identifying transcription factors that modify AR function. We have shown that nuclear factor I/B (NFIB) regulates AR activity in androgen-dependent prostate cancer cells in vitro. However, the status of NFIB in prostate cancer was unknown. METHODS: We immunostained a tissue microarray including normal, hyperplastic, prostatic intraepithelial neoplasia, primary prostatic adenocarcinoma, and castration-resistant prostate cancer tissue samples for NFIB, AR, and synaptophysin, a marker of neuroendocrine differentiation. We interrogated publically available data sets in cBioPortal to correlate NFIB expression and AR and neuroendocrine prostate cancer (NEPCa) activity scores. We analyzed prostate cancer cell lines for NFIB expression via Western blot analysis and used nuclear and cytoplasmic fractionation to assess where NFIB is localized. We performed co-immunoprecipitation studies to determine if NFIB and AR interact. RESULTS: NFIB increased in the nucleus and cytoplasm of prostate cancer samples versus matched normal controls, independent of Gleason score. Similarly, cytoplasmic AR and synaptophysin increased in primary prostate cancer. We observed strong NFIB staining in primary small cell prostate cancer. The ratio of cytoplasmic-to-nuclear NFIB staining was predictive of earlier biochemical recurrence in prostate cancer, once adjusted for tumor margin status. Cytoplasmic AR was an independent predictor of biochemical recurrence. There was no statistically significant difference between NFIB and synaptophysin expression in primary and castration-resistant prostate cancer, but cytoplasmic AR expression was increased in castration-resistant samples. In primary prostate cancer, nuclear NFIB expression correlated with cytoplasmic NFIB and nuclear AR, while cytoplasmic NFIB correlated with synaptophysin, and nuclear and cytoplasmic AR. In castration-resistant prostate cancer samples, NFIB expression correlated positively with an AR activity score, and negatively with the NEPCa score. In prostate cancer cell lines, NFIB exists in several isoforms. We observed NFIB predominantly in the nuclear fraction of prostate cancer cells with increased cytoplasmic expression seen in castration-resistant cell lines. We observed an interaction between AR and NFIB through co-immunoprecipitation experiments. CONCLUSION: We have described the expression pattern of NFIB in primary and castration-resistant prostate cancer and its positive correlation with AR. We have also demonstrated AR interacts with NFIB.


Subject(s)
NFI Transcription Factors/biosynthesis , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Cell Line, Tumor , Gene Expression , Humans , Immunohistochemistry , Male , NFI Transcription Factors/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Tissue Array Analysis , Transcriptome
5.
Prostate ; 80(10): 731-741, 2020 07.
Article in English | MEDLINE | ID: mdl-32356572

ABSTRACT

BACKGROUND: Male lower urinary tract symptoms (LUTS) occur in more than half of men above 50 years of age. LUTS were traditionally attributed to benign prostatic hyperplasia (BPH) and therefore the clinical terminology often uses LUTS and BPH interchangeably. More recently, LUTS were also linked to fibrogenic and inflammatory processes. We tested whether osteopontin (OPN), a proinflammatory and profibrotic molecule, is increased in symptomatic BPH. We also tested whether prostate epithelial and stromal cells secrete OPN in response to proinflammatory stimuli and identified downstream targets of OPN in prostate stromal cells. METHODS: Immunohistochemistry was performed on prostate sections obtained from the transition zone of patients who underwent surgery (Holmium laser enucleation of the prostate) to relieve LUTS (surgical BPH, S-BPH) or patients who underwent radical prostatectomy to remove low-grade prostate cancer (incidental BPH, I-BPH). Images of stained tissue sections were captured with a Nuance Multispectral Imaging System and histoscore, as a measure of OPN staining intensity, was determined with inForm software. OPN protein abundance was determined by Western blot analysis. The ability of prostate cells to secrete osteopontin in response to IL-1ß and TGF-ß1 was determined in stromal (BHPrS-1) and epithelial (NHPrE-1 and BHPrE-1) cells by enzyme-linked immunosorbent assay. Quantitative polymerase chain reaction was used to measure gene expression changes in these cells in response to OPN. RESULTS: OPN immunostaining and protein levels were more abundant in S-BPH than I-BPH. Staining was distributed across all cell types with the highest levels in epithelial cells. Multiple OPN protein variants were identified in immortalized prostate stromal and epithelial cells. TGF-ß1 stimulated OPN secretion by NHPrE-1 cells and both IL-1ß and TGF-ß1 stimulated OPN secretion by BHPrS-1 cells. Interestingly, recombinant OPN increased the mRNA expression of CXCL1, CXCL2, CXCL8, PTGS2, and IL6 in BHPrS-1, but not in epithelial cell lines. CONCLUSIONS: OPN is more abundant in prostates of men with S-BPH compared to men with I-BPH. OPN secretion is stimulated by proinflammatory cytokines, and OPN acts directly on stromal cells to drive the synthesis of proinflammatory mRNAs. Pharmacological manipulation of prostatic OPN may have the potential to reduce LUTS by inhibiting both inflammatory and fibrotic pathways.


Subject(s)
Osteopontin/biosynthesis , Prostatic Hyperplasia/metabolism , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Humans , Immunohistochemistry , Interleukin-6/biosynthesis , Interleukin-6/genetics , Male , Osteopontin/genetics , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stromal Cells/metabolism , Stromal Cells/pathology
6.
Proc Natl Acad Sci U S A ; 114(6): 1359-1364, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28123062

ABSTRACT

The etiology of benign prostatic hyperplasia (BPH) is multifactorial, and chronic inflammation plays a pivotal role in its pathogenesis. Growth hormone-releasing hormone (GHRH) is a hypothalamic neuropeptide that has been shown to act as paracrine/autocrine factor in various malignancies including prostate cancer. GHRH and its receptors are expressed in experimental models of BPH, in which antagonists of GHRH suppressed the levels of proinflammatory cytokines and altered the expression of genes related to epithelial-to-mesenchymal transition (EMT). We investigated the effects of GHRH antagonist on prostatic enlargement induced by inflammation. Autoimmune prostatitis in Balb/C mice was induced by a homogenate of reproductive tissues of male rats. During the 8-wk induction of chronic prostatitis, we detected a progressive increase in prostatic volume reaching 92% at week 8 compared with control (P < 0.001). Daily treatment for 1 mo with GHRH antagonist MIA-690 caused a 30% reduction in prostate volume (P < 0.05). Conditioned medium derived from macrophages increased the average volume of spheres by 82.7% (P < 0.001) and elevated the expression of mRNA for N-cadherin, Snail, and GHRH GHRH antagonist reduced the average volume of spheres stimulated by inflammation by 75.5% (P < 0.05), and TGF-ß2 by 91.8% (P < 0.01). The proliferation of primary epithelial cells stimulated by IL-17A or TGF-ß2 was also inhibited by 124.1% and 69.9%, respectively. GHRH stimulated the growth of BPH-1 and primary prostate spheres. This study provides evidence that GHRH plays important roles in prostatic inflammation and EMT and suggests the merit of further investigation to elucidate the effects of GHRH antagonists in prostatitis and BPH.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression/drug effects , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Humans , Male , Mice, Inbred BALB C , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatitis/genetics , Prostatitis/metabolism , Prostatitis/pathology , Rats , Transforming Growth Factor beta2/pharmacology
7.
Prostate ; 78(13): 970-980, 2018 09.
Article in English | MEDLINE | ID: mdl-29786867

ABSTRACT

BACKGROUND: Inflammation plays a key role in the etiology of benign prostatic hyperplasia (BPH) through multiple pathways involving the stimulation of proliferation by cytokines and growth factors as well as the induction of the focal occurrence of epithelial-to-mesenchymal transition (EMT). We have previously reported that GHRH acts as a prostatic growth factor in experimental BPH and in autoimmune prostatitis models and its blockade with GHRH antagonists offer therapeutic approaches for these conditions. Our current study was aimed at the investigation of the beneficial effects of GHRH antagonists in λ-carrageenan-induced chronic prostatitis and at probing the downstream molecular pathways that are implicated in GHRH signaling. METHODS: To demonstrate the complications triggered by recurrent/chronic prostatic inflammation in Sprague-Dawley rats, 50 µL 3% carrageenan was injected into both ventral prostate lobes two times, 3 weeks apart. GHRH antagonist, MIA-690, was administered 5 days after the second intraprostatic injection at 20 µg daily dose for 4 weeks. GHRH-induced signaling events were identified in BPH-1 and in primary prostate epithelial (PrEp) cells at 5, 15, 30, and 60 min with Western blot. RESULTS: Inflammation induced prostatic enlargement and increased the area of the stromal compartment whereas treatment with the GHRH antagonist significantly reduced these effects. This beneficial activity was consistent with a decrease in prostatic GHRH, inflammatory marker COX-2, growth factor IGF-1 and inflammatory and EMT marker TGF-ß1 protein levels and the expression of multiple genes related to EMT. In vitro, GHRH stimulated multiple pathways involved in inflammation and growth in both BPH-1 and PrEp cells including NFκB p65, AKT, ERK1/2, EGFR, STAT3 and increased the levels of TGF-ß1 and Snail/Slug. Most interestingly, GHRH also stimulated the transactivation of the IGF receptor. CONCLUSIONS: The study demonstrates that GHRH antagonists could be beneficial for the treatment of prostatic inflammation and BPH in part by inhibiting the growth-promoting and inflammatory effects of locally produced GHRH.


Subject(s)
Growth Hormone-Releasing Hormone/antagonists & inhibitors , Prostatic Hyperplasia/drug therapy , Prostatitis/drug therapy , Animals , Carrageenan , Cell Line , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Male , Prostatic Hyperplasia/chemically induced , Prostatic Hyperplasia/metabolism , Prostatitis/chemically induced , Prostatitis/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
8.
Br J Haematol ; 181(4): 476-485, 2018 05.
Article in English | MEDLINE | ID: mdl-29663325

ABSTRACT

Growth hormone-releasing hormone (GHRH) is secreted by the hypothalamus and acts on the pituitary gland to stimulate the release of growth hormone (GH). GHRH can also be produced by human cancers, in which it functions as an autocrine/paracrine growth factor. We have previously shown that synthetic antagonistic analogues of GHRH are able to successfully suppress the growth of 60 different human cancer cell lines representing over 20 cancers. Nevertheless, the expression of GHRH and its receptors in leukaemias has never been examined. Our study demonstrates the presence of GHRH receptor (GHRH-R) on 3 of 4 human acute myeloid leukaemia (AML) cell lines-K-562, THP-1, and KG-1a-and significant inhibition of proliferation of these three cell lines in vitro following incubation with the GHRH antagonist MIA-602. We further show that this inhibition of proliferation is associated with the upregulation of pro-apoptotic genes and inhibition of Akt signalling in leukaemic cells. Treatment with MIA-602 of mice bearing xenografts of these human AML cell lines drastically reduced tumour growth. The expression of GHRH-R was further confirmed in 9 of 9 samples from patients with AML. These findings offer a new therapeutic approach to this malignancy and suggest a possible role of GHRH-R signalling in the pathology of AML.


Subject(s)
Apoptosis/drug effects , Drug Delivery Systems/methods , Leukemia, Myeloid, Acute/drug therapy , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Pituitary Hormone-Regulating Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Signal Transduction/drug effects , Animals , Female , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism , Sermorelin/pharmacology , THP-1 Cells , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 112(44): 13651-6, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26474831

ABSTRACT

Agonists of growth hormone-releasing hormone (GHRH) have been previously reported to promote growth, function, and engraftment of islet cells following transplantation. Here we evaluated recently synthesized GHRH agonists on the proliferation and biological functions of rat pancreatic ß-cell line (INS-1) and islets. In vitro treatment of INS-1 cells with GHRH agonists increased cell proliferation, the expression of cellular insulin, insulin-like growth factor-1 (IGF1), and GHRH receptor, and also stimulated insulin secretion in response to glucose challenge. Exposure of INS-1 cells to GHRH agonists, MR-356 and MR-409, induced activation of ERK and AKT pathways. Agonist MR-409 also significantly increased the levels of cellular cAMP and the phosphorylation of cAMP response element binding protein (CREB) in INS-1 cells. Treatment of rat islets with agonist, MR-409 significantly increased cell proliferation, islet size, and the expression of insulin. In vivo daily s.c. administration of 10 µg MR-409 for 3 wk dramatically reduced the severity of streptozotocin (STZ)-induced diabetes in nonobese diabetic severe combined immunodeficiency (NOD/SCID) mice. The maximal therapeutic benefits with respect to the efficiency of engraftment, ability to reach normoglycemia, gain in body weight, response to high glucose challenge, and induction of higher levels of serum insulin and IGF1 were observed when diabetic mice were transplanted with rat islets preconditioned with GHRH agonist, MR-409, and received additional treatment with MR-409 posttransplantation. This study provides an improved approach to the therapeutic use of GHRH agonists in the treatment of diabetes mellitus.


Subject(s)
Growth Hormone-Releasing Hormone/agonists , Animals , Cell Line, Tumor , Mice , Mice, Inbred NOD , Mice, SCID , Rats , Streptozocin
10.
Proc Natl Acad Sci U S A ; 111(2): 781-6, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24379381

ABSTRACT

The dismal prognosis of malignant brain tumors drives the development of new treatment modalities. In view of the multiple activities of growth hormone-releasing hormone (GHRH), we hypothesized that pretreatment with a GHRH agonist, JI-34, might increase the susceptibility of U-87 MG glioblastoma multiforme (GBM) cells to subsequent treatment with the cytotoxic drug, doxorubicin (DOX). This concept was corroborated by our findings, in vivo, showing that the combination of the GHRH agonist, JI-34, and DOX inhibited the growth of GBM tumors, transplanted into nude mice, more than DOX alone. In vitro, the pretreatment of GBM cells with JI-34 potentiated inhibitory effects of DOX on cell proliferation, diminished cell size and viability, and promoted apoptotic processes, as shown by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide proliferation assay, ApoLive-Glo multiplex assay, and cell volumetric assay. Proteomic studies further revealed that the pretreatment with GHRH agonist evoked differentiation decreasing the expression of the neuroectodermal stem cell antigen, nestin, and up-regulating the glial maturation marker, GFAP. The GHRH agonist also reduced the release of humoral regulators of glial growth, such as FGF basic and TGFß. Proteomic and gene-expression (RT-PCR) studies confirmed the strong proapoptotic activity (increase in p53, decrease in v-myc and Bcl-2) and anti-invasive potential (decrease in integrin α3) of the combination of GHRH agonist and DOX. These findings indicate that the GHRH agonists can potentiate the anticancer activity of the traditional chemotherapeutic drug, DOX, by multiple mechanisms including the induction of differentiation of cancer cells.


Subject(s)
Drug Therapy/methods , Glioblastoma/drug therapy , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/agonists , Peptide Fragments/pharmacology , Animals , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein , Growth Hormone-Releasing Hormone/pharmacology , Immunohistochemistry , Mice , Mice, Nude , Nerve Tissue Proteins/metabolism , Nestin/metabolism , Real-Time Polymerase Chain Reaction
11.
Histochem Cell Biol ; 145(2): 163-73, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26671787

ABSTRACT

Neurite growth is central to the formation and differentiation of functional neurons, and recently, an essential role for phospholipase C-η2 (PLCη2) in neuritogenesis was revealed. Here we investigate the function of PLCη2 in neuritogenesis using Neuro2A cells, which upon stimulation with retinoic acid differentiate and form neurites. We first investigated the role of the PLCη2 calcium-binding EF-hand domain, a domain that is known to be required for PLCη2 activation. To do this, we quantified neurite outgrowth in Neuro2A cells, stably overexpressing wild-type PLCη2 and D256A (EF-hand) and H460Q (active site) PLCη2 mutants. Retinoic acid-induced neuritogenesis was highly dependent on PLCη2 activity, with the H460Q mutant exhibiting a strong dominant-negative effect. Expression of the D256A mutant had little effect on neurite growth relative to the control, suggesting that calcium-directed activation of PLCη2 is not essential to this process. We next investigated which cellular compartments contain endogenous PLCη2 by comparing immunoelectron microscopy signals over control and knockdown cell lines. When signals were analyzed to reveal specific labeling for PLCη2, it was found to be localized predominantly over the nucleus and cytosol. Furthermore in these compartments (and also in growing neurites), a proximity ligand assay revealed that PLCη2 specifically interacts with LIMK-1 in Neuro2A cells. Taken together, these data emphasize the importance of the PLCη2 EF-hand domain and articulation of PLCη2 with LIMK-1 in regulating neuritogenesis.


Subject(s)
Cell Nucleolus/metabolism , Cytoplasm/metabolism , Lim Kinases/metabolism , Neurites/drug effects , Phosphoinositide Phospholipase C/metabolism , Tretinoin/pharmacology , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Nucleolus/chemistry , Cytoplasm/chemistry , Mice , Phosphoinositide Phospholipase C/genetics , Protein Binding
12.
Proc Natl Acad Sci U S A ; 110(36): 14801-6, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23959899

ABSTRACT

Inflammation-related dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis is central to the course of systemic inflammatory response syndrome or sepsis. The underlying mechanisms, however, are not well understood. Initial activation of adrenocortical hormone production during early sepsis depends on the stimulation of hypothalamus and pituitary mediated by cytokines; in late sepsis, there is a shift from neuroendocrine to local immune-adrenal regulation of glucocorticoid production. Therefore, the modulation of the local immune-adrenal cross talk, and not of the neuroendocrine circuits involved in adrenocorticotropic hormone production, may be more promising in the prevention of the adrenal insufficiency associated with prolonged sepsis. In the present work, we investigated the function of the crucial Toll-like receptor (TLR) adaptor protein myeloid differentiation factor 88 (MyD88) in systemic and local activation of adrenal gland inflammation and glucocorticoid production mediated by lipopolysachharides (LPSs). To this end, we used mice with a conditional MyD88 allele. These mice either were interbred with Mx1 Cre mice, resulting in systemic MyD88 deletion, predominantly in the liver and hematopoietic system, or were crossed with Akr1b7 Cre transgenic mice, resulting thereby in deletion of MyD88, which was adrenocortical-specific. Although reduced adrenal inflammation and HPA-axis activation mediated by LPS were found in Mx1(Cre+)-MyD88(fl/fl) mice, adrenocortical-specific MyD88 deletion did not alter the adrenal inflammation or HPA-axis activity under systemic inflammatory response syndrome conditions. Thus, our data suggest an important role of immune cell rather than adrenocortical MyD88 for adrenal inflammation and HPA-axis activation mediated by LPS.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Inflammation/physiopathology , Myeloid Differentiation Factor 88/physiology , Pituitary-Adrenal System/physiology , Adrenal Cortex/cytology , Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Animals , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Female , Gene Expression , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/drug effects , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Toll-Like Receptors/metabolism
13.
J Cell Biochem ; 115(3): 557-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24123053

ABSTRACT

Phospholipase C-η (PLCη) enzymes are a class of phosphatidylinositol 4,5-bisphosphate-hydrolyzing enzymes involved in intracellular signaling. PLCη2 can sense Ca(2+) (stimulated by ∼1 µM free Ca(2+) ) suggesting that it can amplify transient Ca(2+) signals. PLCη enzymes possess an EF-hand domain composed of two EF-loops; a canonical 12-residue loop (EF-loop 1) and a non-canonical 13-residue loop (EF-loop 2). Ca(2+) -binding to synthetic peptides corresponding to EF-loops 1 and 2 of PLCη2 and EF-loop 1 of calmodulin (as a control) was examined by 2D-[(1) H,(1) H] TOCSY NMR. Both PLCη2 EF-loop peptides bound Ca(2+) in a similar manner to that of the canonical calmodulin EF-loop 1, particularly at their N-terminus. A molecular model of the PLCη2 EF-hand domain, constructed based upon the structure of calmodulin, suggested both EF-loops may participate in Ca(2+) -binding. To determine whether the EF-hand is responsible for Ca(2+) -sensing, inositol phosphate accumulation was measured in COS7 cells transiently expressing wild-type or mutant PLCη2 proteins. Addition of 70 µM monensin (a Na(+) /H(+) antiporter that increases intracellular Ca(2+) ) induced a 4- to 7-fold increase in wild-type PLCη2 activity. In permeabilized cells, PLCη2 exhibited a ∼4-fold increase in activity in the presence of 1 µM free Ca(2+) . The D256A (EF-loop1) mutant exhibited a ∼10-fold reduction in Ca(2+) -sensitivity and was not activated by monensin, highlighting the involvement of EF-loop 1 in Ca(2+) -sensing. Involvement of EF-loop 2 was examined using D292A, H296A, Q297A, and E304A mutants. Interestingly, the monensin responses and Ca(2+) -sensitivities were largely unaffected by the mutations, indicating that the non-canonical EF-loop 2 is not involved in Ca(2+) -sensing.


Subject(s)
Calcium/metabolism , Models, Molecular , Phosphoinositide Phospholipase C/chemistry , Protein Conformation , Amino Acid Sequence , Animals , COS Cells , Calcium/chemistry , Calmodulin/chemistry , Chlorocebus aethiops , EF Hand Motifs/genetics , Humans , Inositol/pharmacology , Mutation/genetics , Phosphoinositide Phospholipase C/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Signal Transduction , Structure-Activity Relationship
14.
bioRxiv ; 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38712029

ABSTRACT

Benign prostatic hyperplasia (BPH) is a prevalent age-related condition often characterized by debilitating urinary symptoms. Its etiology is believed to stem from hormonal imbalance, particularly an elevated estradiol-to-testosterone ratio and chronic inflammation. Our previous studies using a mouse steroid hormone imbalance model identified a specific increase in macrophages that migrate and accumulate in the prostate lumen where they differentiate into lipid-laden foam cells in mice implanted with testosterone and estradiol pellets, but not in sham animals. The current study focused on further characterizing the cellular heterogeneity of the prostate in this model as well as identifying the specific transcriptomic signature of the recruited foam cells. Moreover, we aimed to identify the epithelia-derived signals that drive macrophage infiltration and luminal translocation. Male C57BL/6J mice were implanted with slow-release testosterone and estradiol pellets (T+E2) and harvested the ventral prostates two weeks later for scRNA-seq analysis, or performed sham surgery. We identified Ear2+ and Cd72+ macrophages that were elevated in response to steroid hormone imbalance, whereas a Mrc1+ resident macrophage population did not change. In addition, an Spp1+ foam cell cluster was almost exclusively found in T+E2 mice. Further markers of foam cells were also identified, including Gpnmb and Trem2, and GPNMB was confirmed as a novel histological marker with immunohistochemistry. Foam cells were also shown to express known pathological factors Vegf, Tgfb1, Ccl6, Cxcl16 and Mmp12. Intriguingly, a screen for chemokines identified the upregulation of epithelial-derived Cxcl17, a known monocyte attractant, in T+E2 prostates suggesting that it might be responsible for the elevated macrophage number as well as their translocation to the lumen. Our study identified macrophage subsets that respond to steroid hormone imbalance as well as further confirmed a potential pathological role of luminal foam cells in the prostate. These results underscore a pathological role of the identified prostate foam cells and suggests CXCL17-mediated macrophage migration as a critical initiating event.

15.
J Neurochem ; 124(5): 632-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23237262

ABSTRACT

Phospholipase C-η2 is a recently identified phospholipase C (PLC) implicated in the regulation of neuronal differentiation/maturation. PLCη2 activity is triggered by intracellular calcium mobilization and likely serves to amplify Ca²âº signals by stimulating further Ca²âº release from Ins(1,4,5)P3-sensitive stores. The role of PLCη2 in neuritogenesis was assessed during retinoic acid (RA)-induced Neuro2A cell differentiation. PLCη2 expression increased two-fold during a 4-day differentiation period. Stable expression of PLCη2-targetted shRNA led to a decrease in the number of differentiated cells and total length of neurites following RA-treatment. Furthermore, RA response element activation was perturbed by PLCη2 knockdown. Using a bacterial two-hybrid screen, we identified LIM domain kinase 1 (LIMK1) as a putative interaction partner of PLCη2. Immunostaining of PLCη2 revealed significant co-localization with LIMK1 in the nucleus and growing neurites in Neuro2A cells. RA-induced phosphorylation of LIMK1 and cAMP-responsive element-binding protein was reduced in PLCη2 knock-down cells. The phosphoinositide-binding properties of the PLCη2 PH domain, assessed using a FRET-based assay, revealed this domain to possess a high affinity toward PtdIns(3,4,5)P3. Immunostaining of PLCη2 together with PtdIns(3,4,5)P3 in the Neuro2A cells revealed a high degree of co-localization, indicating that PtdIns(3,4,5)P3 levels in cellular compartments are likely to be important for the spatial control of PLCη2 signaling.


Subject(s)
Neurites/metabolism , Neurons/cytology , Neurons/metabolism , Phosphoinositide Phospholipase C/metabolism , Tretinoin/pharmacology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/physiology , Lim Kinases/metabolism , Mice , Neurites/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Biomedicines ; 11(11)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-38001899

ABSTRACT

The prostate gland, located beneath the bladder and surrounding the proximal urethra in men, plays a vital role in reproductive physiology and sexual health. Despite its importance, the prostate is vulnerable to various pathologies, including prostatitis, benign prostatic hyperplasia (BPH) and prostate cancer (PCa). Osteopontin (OPN), a versatile protein involved in wound healing, inflammatory responses, and fibrotic diseases, has been implicated in all three prostate conditions. The role of OPN in prostatic pathophysiology, affecting both benign and malignant prostate conditions, is significant. Current evidence strongly suggests that OPN is expressed at a higher level in prostate cancer and promotes tumor progression and aggressiveness. Conversely, OPN is primarily secreted by macrophages and foam cells in benign prostate conditions and provokes inflammation and fibrosis. This review discusses the accumulating evidence on the role of OPN in prostatic diseases, cellular sources, and potential roles while also highlighting areas for future investigations.

17.
Biochem Soc Trans ; 40(1): 282-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22260706

ABSTRACT

The most recently identified PLC (phospholipase C) enzymes belong to the PLCη family. Their unique Ca2+-sensitivity and their specific appearance in neurons have attracted great attention since their discovery; however, their physiological role(s) in neurons are still yet to be established. PLCη enzymes are expressed in the neocortex, hippocampus and cerebellum. PLCη2 is also expressed at high levels in pituitary gland, pineal gland and in the retina. Driven by the specific localization of PLCη enzymes in different brain areas, in the present paper, we discuss the roles that they may play in neural processes, including differentiation, memory formation, circadian rhythm regulation, neurotransmitter/hormone release and the pathogenesis of neurodegenerative disorders associated with aberrant Ca2+ signalling, such as Alzheimer's disease.


Subject(s)
Calcium Signaling , Neurons/physiology , Synaptic Transmission , Type C Phospholipases/physiology , Animals , Brain/cytology , Brain/enzymology , Brain/physiology , Humans , Neurogenesis , Neurons/enzymology , Neurons/metabolism , Synapses/enzymology , Synapses/metabolism , Type C Phospholipases/metabolism
18.
Cell Mol Life Sci ; 68(1): 85-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20812023

ABSTRACT

GPR39 is a vertebrate G protein-coupled receptor related to the ghrelin/neurotensin receptor subfamily. The receptor is expressed in a range of tissues including the pancreas, gut/gastrointestinal tract, liver, kidney and in some regions of the brain. GPR39 was initially thought to be the cognitive receptor for the peptide hormone, obestatin. However, subsequent in vitro studies have failed to demonstrate binding of this peptide to the receptor. Zn(2+) has been shown to be a potent stimulator of GPR39 activity via the Gα(q), Gα(12/13) and Gα(s) pathways. The potency and specificity of Zn(2+) in activating GPR39 suggest it to be a physiologically important agonist. GPR39 is now emerging as an important transducer of autocrine and paracrine Zn(2+) signals, impacting upon cellular processes such as insulin secretion, gastric emptying, neurotransmission and epithelial repair. This review focuses on the molecular, structural and biological properties of GPR39 and its various physiological functions.


Subject(s)
Gastrointestinal Tract/physiology , Neurons/physiology , Pancreas/physiology , Receptors, G-Protein-Coupled/physiology , Zinc/metabolism , Amino Acid Sequence , Animals , Cattle , Gastrointestinal Tract/metabolism , Humans , Mice , Molecular Sequence Data , Neurons/metabolism , Pancreas/metabolism , Protein Structure, Tertiary , Rats , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Sequence Alignment , Signal Transduction , Wound Healing
19.
Am J Clin Exp Urol ; 10(6): 449-461, 2022.
Article in English | MEDLINE | ID: mdl-36636691

ABSTRACT

The Annual Collaborating for the Advancement of Interdisciplinary Research (CAIRIBU) Meeting in 2022 highlighted basic, translational, and clinical non-malignant urology research within five main areas affecting the urinary tract: urinary dysfunction due to prostate disease, microbes and infection, bladder function and physiology, neurology and neuromuscular influences and calculi and obstruction. In this paper, we summarize main findings and future directions outlined by CAIRIBU-affiliated scientists who presented as part of the scientific sessions.

20.
Peptides ; 150: 170716, 2022 04.
Article in English | MEDLINE | ID: mdl-34952135

ABSTRACT

The syntheses and biological evaluation of GHRH antagonists of AVR series with high anticancer and anti-inflammatory activities are described. Compared to our previously reported GHRH antagonist 602 of MIAMI series, AVR analogs contain additional modifications at positions 0, 6, 8, 10, 11, 12, 20, 21, 29 and 30, which induce greater antitumor activities. Five of nineteen tested AVR analogs presented binding affinities to the membrane GHRH receptors on human pituitary, 2-4-fold better than MIA-602. The antineoplastic properties of these analogs were evaluated in vitro using proliferation assays and in vivo in nude mice xenografted with various human cancer cell lines including lung (NSCLC-ADC HCC827 and NSCLC H460), gastric (NCI-N87), pancreatic (PANC-1 and CFPAC-1), colorectal (HT-29), breast (MX-1), glioblastoma (U87), ovarian (SK-OV-3 and OVCAR-3) and prostatic (PC3) cancers. In vitro AVR analogs showed inhibition of cell viability equal to or greater than MIA-602. After subcutaneous administration at 5 µg/day doses, some AVR antagonists demonstrated better inhibition of tumor growth in nude mice bearing various human cancers, with analog AVR-353 inducing stronger suppression than MIA-602 in lung, gastric, pancreatic and colorectal cancers and AVR-352 in ovarian cancers and glioblastoma. Both antagonists induced greater inhibition of GH release than MIA-602 in vitro in cultured rat pituitary cells and in vivo in rats. AVR-352 also demonstrated stronger anti-inflammatory effects in lung granulomas from mice with lung inflammation. Our studies demonstrate the merit of further investigation of AVR GHRH antagonists and support their potential use for clinical therapy of human cancers and other diseases.


Subject(s)
Glioblastoma , Lung Neoplasms , Ovarian Neoplasms , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Cell Line, Tumor , Female , Growth Hormone , Growth Hormone-Releasing Hormone , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Rats , Sermorelin/metabolism , Sermorelin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL