Elsevier

Differentiation

Volume 82, Issues 4–5, November–December 2011, Pages 200-210
Differentiation

Invited Review
The functional role of reactive stroma in benign prostatic hyperplasia

https://doi.org/10.1016/j.diff.2011.05.007Get rights and content

Abstract

The human prostate gland is one of the only internal organs that continue to enlarge throughout adulthood. The specific mechanisms that regulate this growth, as well as the pathological changes leading to the phenotype observed in the disease benign prostatic hyperplasia (BPH), are essentially unknown. Recent studies and their associated findings have made clear that many complex alterations occur, involving persistent and chronic inflammation, circulating hormonal level deregulation, and aberrant wound repair processes. BPH has been etiologically characterized as a progressive, albeit discontinuous, hyperplasia of both the glandular epithelial and the stromal cell compartments coordinately yielding an expansion of the prostate gland and clinical symptoms. Interestingly, the inflammatory and repair responses observed in BPH are also key components of general wound repair in post-natal tissues. These responses include altered expression of chemokines, cytokines, matrix remodeling factors, chronic inflammatory processes, altered immune surveillance and recognition, as well as the formation of a prototypical ‘reactive’ stroma, which is similar to that observed across various fibroplasias and malignancies of a variety of tissue sites. Stromal tissue, both embryonic mesenchyme and adult reactive stroma myofibroblasts, has been shown to exert potent and functional regulatory control over epithelial proliferation and differentiation as well as immunoresponsive modulation. Thus, the functional biology of a reactive stroma, within the context of an adult disease typified by epithelial and stromal aberrant hyperplasia, is critical to understand within the context of prostate disease and beyond. The mechanisms that regulate reactive stroma biology in BPH represent targets of opportunity for new therapeutic approaches that may extend to other tissue contexts. Accordingly, this review seeks to address the dissection of important factors, signaling pathways, genes, and other regulatory components that mediate the interplay between epithelium and stromal responses in BPH.

Section snippets

Benign prostatic hyperplasia and the reactive phenotype

The human prostate gland is composed of secretory epithelium arranged in glandular acini within a fibromuscular stroma composed primarily of smooth muscle. The stromal compartment also contains fibroblasts, vasculature, nerves and immune components. In an interactive manner, each of these epithelial and stromal components is likely involved in the genesis and evolution of benign prostatic hyperplasia (BPH). Understanding prostate gland development is helpful for interpreting some of the

Epithelial barrier function and the reactive stroma response

Epithelia, though specialized for unique functions throughout the body, share several common morphological features. In general, epithelia line a given surface, directly apposed one to another, yielding a layer. Epithelial tight junctions and other junctional complexes allow for a tight and fairly robust structural integrity of this epithelial lining layer, thus protecting against frictional expansion and contraction within dynamic exocrine organs, such as the prostate. Moreover, the apical

Reactive stroma repair and myofibroblasts

Although the mechanisms of genesis and/or recruitment of myofibroblasts to diseased tissue regions are as yet unresolved (Phan, 2008), myofibroblasts play a central role in fibrotic diseases of the skin, liver, pancreas, kidney and urogenital tissues, among others. Myofibroblasts exhibit properties of both smooth muscle cells and fibroblasts, secreting a host of growth factors and chemokines (Powell et al., 1999), and are defined here histologically as co-expressing vimentin, smooth muscle α

Benign prostatic hyperplasia and chronic inflammation

At present, there is no consensus on the etiology of BPH. There have been many suggestions, some mechanical in nature, such as altered urodynamic function due to increased prostatic urethral angulation (Cho et al., 2008). Some have identified potential molecular events gone awry, such as elevated oxidative stress (Gradini et al., 1999, Aryal et al., 2007), ischemic damage due to vascular impairment (Berger et al., 2005), loss of negative regulators of cell cycle control (Cordon-Cardo et al.,

Chemokines and cytokines in BPH reactive stroma

Many chemokines and cytokines have been shown to be associated with BPH. The CXC chemokines shown to be specifically associated with BPH include IL-1α (Giri and Ittmann, 2000), IL-2 (Royuela et al., 2000), IL-8 (Penna et al., 2007, Castro et al., 2004), IL-15 and IL-17 (Handisurya et al., 2001, Steiner et al., 2002). Among these, IL-8 was shown to be expressed specifically in BPH epithelial cells, and to stimulate expression of fibroblast growth factor 2 (FGF2) in vitro (Castro et al., 2004,

Interleukin-8 and benign prostatic hyperplasia

Interleukins, in general, are mediators of inflammatory processes and are central to induction of wound repair responses and have been implicated in fibrosis. In the prostate gland, IL-8, IL-1α and IL-6 have each gained attention. Overexpression of IL-8 is universally observed in many proliferative disorders and sites of inflammation. IL-8 is a multifunctional chemokine interleukin that regulates many immune host responses and wound repair mechanisms. The elevated expression of several

Extracellular matrix in reactive stroma and BPH

Tenascin-C is an extracellular matrix glycoprotein that exhibits altered expression and deposition patterns in human BPH. We have shown that this is statistically correlated with elevated epithelial expression of IL-8 (Schauer et al., 2008). Moreover, tenascin-C is deposited in activated fibroblasts/myofibroblasts immediately adjacent to BPH epithelial acini. These cells have a phenotype very similar to what we have reported for reactive stroma in prostate cancer. Reactive stroma consisting of

Summary and conclusions

The etiology and specific mechanisms that lead to phenotypic changes that manifest as benign prostate disease remain poorly understood. Recent data suggest that pathophysiological signaling mechanisms are complex, likely involving age-related and chronic defects in tissue homeostasis that lead to compensatory and reactive changes in both the stroma and the epithelial tissue compartments. The historical perspective of BPH biology suggests that the compensatory biology likely involves

References (153)

  • J. Campisi

    Cellular senescence: putting the paradoxes in perspective

    Curr. Opin. Genet. Dev.

    (2011)
  • M. Chiquet et al.

    Induction of tenascin-C by cyclic tensile strain versus growth factors: distinct contributions by Rho/ROCK and MAPK signaling pathways

    Biochim. Biophys. Acta

    (2004)
  • K.S. Cho

    The overlooked cause of benign prostatic hyperplasia: prostatic urethral angulation

    Med. Hypotheses

    (2008)
  • N.B. Delongchamps

    Evaluation of prostatitis in autopsied prostates—is chronic inflammation more associated with benign prostatic hyperplasia or cancer?

    J. Urol.

    (2008)
  • R.M. Devalaraja

    Delayed wound healing in CXCR2 knockout mice

    J. Invest. Dermatol.

    (2000)
  • F. Di Silverio

    Distribution of inflammation, pre-malignant lesions, incidental carcinoma in histologically confirmed benign prostatic hyperplasia: a retrospective analysis

    Eur. Urol.

    (2003)
  • S.M. Eck

    Matrix metalloproteinase and G protein coupled receptors: co-conspirators in the pathogenesis of autoimmune disease and cancer

    J. Autoimmun.

    (2009)
  • K. Fujinaga

    Locally applied cilostazol suppresses neointimal hyperplasia by inhibiting tenascin-C synthesis and smooth muscle cell proliferation in free artery grafts

    J. Thorac. Cardiovasc. Surg.

    (2004)
  • D. Giri et al.

    Interleukin-1alpha is a paracrine inducer of FGF7, a key epithelial growth factor in benign prostatic hyperplasia

    Am. J. Pathol.

    (2000)
  • D. Giri et al.

    Interleukin-8 is a paracrine inducer of fibroblast growth factor 2, a stromal and epithelial growth factor in benign prostatic hyperplasia

    Am. J. Pathol.

    (2001)
  • J. Heidemann

    Angiogenic effects of interleukin 8 (CXCL8) in human intestinal microvascular endothelial cells are mediated by CXCR2

    J. Biol. Chem.

    (2003)
  • K.C. Ingham et al.

    Localization of a cryptic binding site for tenascin on fibronectin

    J. Biol. Chem.

    (2004)
  • K. Ishii

    Role of stromal tenascin-C in mouse prostatic development and epithelial cell differentiation

    Dev. Biol.

    (2008)
  • J.H. Jang et al.

    Tenascin-C promotes cell survival by activation of Akt in human chondrosarcoma cell

    Cancer Lett.

    (2005)
  • J.H. Jang

    Identification and kinetics analysis of a novel heparin-binding site (KEDK) in human tenascin-C

    J. Biol. Chem.

    (2004)
  • Y.D. Jung

    Role of P38 MAPK, AP-1, and NF-kappaB in interleukin-1beta-induced IL-8 expression in human vascular smooth muscle cells

    Cytokine

    (2002)
  • S.J. Kim

    Expression of interleukin-8 correlates with angiogenesis, tumorigenicity, and metastasis of human prostate cancer cells implanted orthotopically in nude mice

    Neoplasia

    (2001)
  • O. Klezovitch

    Hepsin promotes prostate cancer progression and metastasis

    Cancer Cell

    (2004)
  • P.W. Kohnen et al.

    Patterns of inflammation in prostatic hyperplasia: a histologic and bacteriologic study

    J. Urol.

    (1979)
  • R. Kooijman

    Regulation of interleukin-8 expression in human prostate cancer cells by insulin-like growth factor-I and inflammatory cytokines

    Growth Horm. IGF Res.

    (2007)
  • Y. Koyama

    Effect of tenascin-C deficiency on chemically induced dermatitis in the mouse

    J. Invest. Dermatol.

    (1998)
  • G. Kramer et al.

    Is benign prostatic hyperplasia (BPH) an immune inflammatory disease?

    Eur. Urol.

    (2007)
  • K.L. Lee et al.

    Molecular and cellular pathogenesis of benign prostatic hyperplasia

    J. Urol.

    (2004)
  • D.Q. Li

    JNK and ERK MAP kinases mediate induction of IL-1beta, TNF-alpha and IL-8 following hyperosmolar stress in human limbal epithelial cells

    Exp. Eye Res.

    (2006)
  • Q. Li

    Matrilysin shedding of syndecan-1 regulates chemokine mobilization and transepithelial efflux of neutrophils in acute lung injury

    Cell

    (2002)
  • I. Malgorzata Goczalik

    The activation of IL-8 receptors in cultured guinea pig Muller glial cells is modified by signals from retinal pigment epithelium

    J. Neuroimmunol.

    (2005)
  • J. McNeal

    Pathology of benign prostatic hyperplasia. Insight into etiology

    Urol. Clin. North Am.

    (1990)
  • C.L. Addison

    The CXC chemokine receptor 2, CXCR2, is the putative receptor for ELR+CXC chemokine-induced angiogenic activity

    J. Immunol.

    (2000)
  • S. Araki

    Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer

    Cancer Res.

    (2007)
  • M. Aryal

    Oxidative stress in patients with benign prostate hyperplasia

    JNMA J. Nepal Med. Assoc.

    (2007)
  • G. Ayala

    Reactive stroma as a predictor of biochemical-free recurrence in prostate cancer

    Clin. Cancer Res.

    (2003)
  • L. Begley

    CXCL12 overexpression and secretion by aging fibroblasts enhance human prostate epithelial proliferation in vitro

    Aging Cell

    (2005)
  • J.A. Belperio

    CXC chemokines in angiogenesis

    J. Leukoc. Biol.

    (2000)
  • A.P. Berger

    Vascular damage as a risk factor for benign prostatic hyperplasia and erectile dysfunction

    BJU Int.

    (2005)
  • D. Bianchi-Frias

    The effects of aging on the molecular and cellular composition of the prostate microenvironment

    PLoS. One

    (2010)
  • K.R. Buchholz et al.

    The extracellular signal-regulated kinase/mitogen-activated protein kinase pathway induces the inflammatory factor interleukin-8 following Chlamydia trachomatis infection

    Infect. Immun.

    (2007)
  • Cantini, L.P., et al., Profibrotic role of myostatin in Peyronie’s disease. J. Sex. Med.,...
  • P. Castro

    Interleukin-8 expression is increased in senescent prostatic epithelial cells and promotes the development of benign prostatic hyperplasia

    Prostate

    (2004)
  • B. Chatterjee

    The role of the androgen receptor in the development of prostatic hyperplasia and prostate cancer

    Mol. Cell. Biochem.

    (2003)
  • S. Claus

    Immunohistochemical determination of age related proliferation rates in normal and benign hyperplastic human prostates

    Urol. Res.

    (1993)
  • Cited by (107)

    • The cancer-inhibitory effects of proliferating tumor-residing fibroblasts

      2022, Biochimica et Biophysica Acta - Reviews on Cancer
    • Serum testosterone and prostate-specific antigen levels are major risk factors for prostatic volume increase among benign prostatic hyperplasia patients

      2021, Asian Journal of Urology
      Citation Excerpt :

      Thus, the independent association of the cytokine solely with prostate volume is difficult to be proven significant as seen in the results of the study. Physiologically, unlike other organs the prostate keeps growing throughout the adult male life [41]. An ample amount of studies in the past have explored the role of testosterone in the increase of prostate volume.

    View all citing articles on Scopus

    Funding Sources: Supported by NIH Grants R01 DK083293 and R01 CA58093.

    View full text