Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 1.
Published in final edited form as: Sex Med Rev. 2016 Jan 1;4(1):85–94. doi: 10.1016/j.sxmr.2015.10.002

The Genetic Basis of Peyronie’s Disease: A Review

Amin S Herati 1,2, Alexander W Pastuszak 1,2
PMCID: PMC4778255  NIHMSID: NIHMS745083  PMID: 27872008

Abstract

Introduction

Peyronie’s disease (PD) is progressive fibrotic disorder of the penile tunica albuginea that results in fibrotic penile plaques and may lead to penile deformity. Characterized by aberrant fibrosis resulting in part from persistence of myofibroblasts as well as altered gene expression, the molecular factors underpinning Peyronie’s disease and other related fibrotic diatheses are just being elucidated. A genetic link to PD was first identified using pedigree analyses three decades ago. However, the specific genetic factors that predispose patients to aberrant fibrosis remain unknown, and the relationships between these fibrotic conditions and other heritable diseases, including malignancy, are uncharacterized.

Aim

To review the current landscape linking molecular and genetic factors to aberrant fibrosis in PD as well as related fibrotic diatheses, including Dupuytren’s disease.

Methods

Review and evaluation of the literature from 1970 to present examining the genetic factors associated with PD.

Main Outcome Measures

Data describing the genetic factors associated with PD.

Results

We describe the known structural chromosomal abnormalities and single nucleotide polymorphisms associated with fibrotic diatheses, and discuss the spectrum of differential gene expression data comparing normal tissues and those derived from men with Peyronie’s or Dupuytren’s diseases. Finally, we discuss epigenetic mechanisms that may regulate gene expression and alter predisposition to fibrosis.

Conclusion

Although our current understanding of the genetic factors associated with PD are limited, significant advances have been made over the last three decades. Further research is necessary to provide a more comprehensive understanding of the landscape of genetic factors responsible for the development of PD.

Keywords: Adult, cell differentiation/genetics, cell division/genetics, fibroblast/pathology, Gene Expression Regulation/Physiology, Humans, Male, Oligonucleotide Array Sequence Analysis, Penile Induration/genetics*, Penile induration/pathology, penile induration/therapy, Transforming Growth Factor beta/genetics, Transforming Growth Factor Beta1

Introduction

Peyronie’s disease (PD) is a progressive fibrotic disorder of the tunica albuginea (TA) of the penis that is defined by the presence of tunical plaques. Clinically, these plaques can result in penile pain and penile deformity including curvature, shortening, and narrowing.1 PD plaques are thought to develop following either acute trauma or repeated external stress or microtrauma to the penis, causing delamination of the bilaminar TA. This initiates the inflammatory cascade and results in an abnormal persistence of myofibroblasts, leading to aberrant collagen accumulation, fibrin deposition, and disordered elastic fibers, ultimately resulting in tunical plaque formation and scarring.26

PD may be a localized manifestation of a systemic fibrotic disorder existing in the same spectrum as Dupuytren’s disease (DD), which affects the palmar fascia of the hand, and Ledderhose disease (LD), which affects the plantar aponeurosis of the foot.79 Concurrent fibrotic syndromes can be seen within the same individual, with 10–20% of men with PD also manifesting DD.7

Although the complete spectrum of etiologic factors for PD is unknown, multiple mechanisms have been proposed including trauma, impaired fibrin clearance, autoimmune and genetic factors.6, 1013 The evidence supporting a genetic predisposition in persons with PD has since been recognized through HLA associations, pedigree analyses confirming a familial inheritance pattern consistent with an autosomal dominant mode of transmission, and gene expression studies supporting a common pathogenetic mechanism.12, 16 In this review, we summarize the literature evaluating genetic factors involved in fibrotic diatheses, with a focus on PD. It is our goal to provide an overview of the known genetic factors that may predispose to aberrant fibrosis, highlighting opportunities for investigation in this relatively unexplored space.

Epidemiology

PD is reported to have an incidence of 22.4 to 25.7 per 100,000 men, with the highest incidence in men 50 to 59 years old; the average age of presentation is 55 years old.17, 18 Despite the higher incidence in older men, PD can affect men of all ages, from teenagers to men over 70 years old.1921 Reported prevalence rates of PD range from 0.4% to 13%, although these are likely underestimates due to an under-reporting bias; men are often reluctant to discuss this condition.21, 22

Although the roles of certain environmental risk factors in the development of PD, including smoking, obesity, and hypertension, have been determined, the epidemiologic impact of genetic predisposition to PD and other fibrotic diatheses remains unknown, largely because genetic causes of PD remain to be determined.23, 24

Familial Aggregation and Genetic Transmission Modes

The initial indication that genetic factors may play a role in the pathogenesis of PD was demonstrated in 1982 by Bias et al.12 through pedigree analysis of three families affected by both PD and DD. An autosomal dominant mode of inheritance with incomplete penetrance was identified in the three families, with one family exhibiting three generations of father-to-son transmission. The presence of a low frequency of affected family members despite the autosomal dominant transmission was later demonstrated by Nugteren et al.7 who showed that of the 415 men in their study with PD, 89 (22.1%) also had DD, 5 (1.4%) had an affected father or brother, and 28 (6.7%) had a positive family history for DD.

Expanding on the finding of Willscher et al.,16 who had previously shown an increased frequency of the HLA-B7 cross-reacting group in patients with idiopathic PD compared to the general population, Bias et al.12 found that antigens of the HLA-B7 cross-reacting group were present among family members of all three families affected by PD. However, the familial segregation patterns suggested independent assortment of HLA loci, decreasing the likelihood that these loci contribute to the development of PD. Three studies analyzing the population frequencies of HLA antigens in the PD population have subsequently debunked the association between HLA antigen expression and PD.2527

Chromosomal Abnormalities in Peyronie’s Disease

Several fibroblast cell culture models have documented chromosomal abnormalities associated with PD. Early work by Somers et al28 detected karyotypic abnormalities in PD plaque-derived fibroblasts in 7 of 12 PD patients. Chromosomal abnormalities detected include three numerical changes including duplication of chromosome 7 and 8 and deletion of chromosome Y in three patients. Structural chromosomal alterations included reciprocal translocations of 46XY,t(11;12)(q11,p11) and 46XY,t(1;5)(q25;q11), as well as inversion of 46XY,inv(7)(p22q36). These chromosomal abnormalities, importantly, were not present in cell cultures derived from adjacent TA, dermis and lymphocytes in men with PD, arguing against the presence of a heritable mutation in these men, but supporting genomic alterations in affected cells. In a series of fourteen patients with PD by Guerneri et al.,29 plaque-derived cells from 9 were similarly found to have structural and numerical chromosomal abnormalities. Eight of the nine patients had aneusomies of the Y chromosome and three also had chromosomal translocations. Interestingly, structural rearrangements were observed during metaphase in two cases, suggesting the possibility of clonal evolution. Although the significance of these karyotypic changes remains to be determined, these numerical and structural changes may influence cell cycle regulation.

Mulhall and colleagues30 demonstrated the presence of clonal evolution in fibroblasts of both PD plaque-derived fibroblasts as well as fibroblasts from uninvolved TA, supporting a potential field defect of the TA in PD. The temporal progression of the chromosomal abnormalities was demonstrated using fluorescent in situ hybridization (FISH) probes for chromosomes 7, 8, 17, 18, X, and Y. As plaque and nonplaque derived fibroblasts grew in culture, they developed morphologic transformations (cell rounding and increased cytoplasm) as well as cytogenetic abnormalities. The most common chromosomal abnormalities detected were aneuploidy of chromosome 7 and 8, followed by chromosome 17 and 18 and lastly the Y and X chromosomes. Plaque-derived fibroblasts demonstrated chromosomal instability during early passages with four of five cell culture lines demonstrating aneusomy as early as the 3rd passage. These findings were in contrast to those in nonplaque-derived fibroblasts, which developed aneuploidy during later passages, albeit with the same pattern of aneuploidy observed in the plaque-derived fibroblasts. Overlapping patterns of chromosomal aberrations were identified in this study between fibroblasts from PD and DD lesions, including trisomy 7 and 8 and deletion of the Y chromosome, suggesting a shared pathway for aberrant fibrosis.

Single Nucleotide Polymorphisms Associated with PD and DD

Elevated levels of transforming growth factor beta 1 (TGFβ1) have previously been described in PD.11. The role of TGFβ1 in PD pathogenesis is supported by induction of PD in the rat model using recombinant TGFβ1 injected into the rat phallus, and the regression of fibrotic plaques via inhibition of the TGFβ1 type 1 receptor using a small molecule inhibitor of activin receptor-like kinase, which normally activates the TGFβ1 type 1 receptor via transphosphorylation.3437 Although the mechanism for elevated levels of TGFβ1 has not been elucidated, it is hypothesized that this increased expression is partly due to the presence of heritable single nucleotide polymorphisms (SNPs) that may impact gene expression. Several SNPs have been identified within the TGFβ1 gene, including rs1800469 (C–509T), rs1800471 (G915C), and rs1982073 (T+29C). Of these polymorphisms, only G915C has been associated with PD. The G915C SNP, which results in substitution of arginine to proline at position 25 in the TGFβ1 protein,42 was shown in a prospective comparison of 111 men with PD to 100 healthy controls to have a significantly higher frequency in men with PD (p=0.04).43

A genome wide association study (GWAS) of 2,325 patients with DD revealed nine chromosomal loci associated with DD susceptibility.44 Eleven SNPs showed significant associations with these chromosomal loci, and of these, six were associated with the wingless-type MMTV integration site (WNT) signaling pathway. In order to determine whether these SNPs were also present in men with PD, allele frequencies for the SNPs were assessed among 111 men with PD and compared to 490 unaffected controls.45 Of the SNPs evaluated, a statistically significant association was observed with SNP rs4730775 at the WNT2 locus on chromosome 7. Taken together, these studies further implicate a common set of genetic factors for both PD and DD.

Gene Expression Profiles

The advent of microarray platforms and microarray gene expression profiling has permitted rapid analysis of gene expression on a genome-wide scale. Such differential gene expression pattern analysis has facilitated characterization of gene expression disparities between normal and pathologic tissue samples, and has also helped characterize molecular pathways, furthering our understanding of disease progression and identifying novel therapeutic targets for numerous diseases.46

Differences in Gene Expression Between PD Lesions and Normal Tunica Albuginea

Some of the first studies evaluating gene expression in men with PD compared PD plaque tissue to either normal TA from control patients or unaffected TA from the same patient. Initial work by Magee et al. compared gene expression profiles between plaque tissue derived from 7 patients with PD and TA obtained from 5 unaffected patients.47 Two complementary but non-overlapping microarray platforms, a Clontech DNA Microarray (Clontech, Palo Alto, California) and an Affymetrix DNA Microarray (Affymetrix, Santa Carla, California), were compared to identify a cost-effective approach to microarray analysis. The Clontech microarray, which analyzes functionally related families of genes, identified 8 up-regulated and 4 down-regulated genes that exhibited at least 2-fold relative difference in gene expression after normalization to global gene expression. The gene with highest level of differential expression in the PD plaque was pleiotrophin (PTN/OSF-1), a growth factor that induces fibroblast proliferation and osteoblast recruitment and osteogenesis (Table 1).48 PTN/OSF-1 overexpression in postnatal bone has the dual effect of promoting differentiation of human bone marrow stromal cells to chondroblasts and inducing a higher bone mineral content.49, 50 Intriguingly, calcification occurs in a subset of PD plaques, although why it occurs in one plaque and not another is currently unknown. The above data support differential gene expression profiles that likely result in variable predisposition to plaque calcification in some men with PD.

Table 1.

Genes with involvement in Peyronie’s and Dupuytren’s diseases

Gene Gene Symbol Chromosomal
Location
Gene Function Reference
Matrix metalloproteinase 2 MMP 2 16q12.2 Breakdown of extracellular
matrix
88
Matrix metalloproteinase 9 MMP 9 20q13.12 Breakdown of extracellular
matrix
88
Thymosin beta-10 TMSB-10 2p11.2 Prevents spontaneous
globular actin monomer
polymerization
88
Thymosin beta-4 TMSB-4 Xq21.3-q22 Actin sequestering protein 88
Cortactin; amplaxin CTTN 11q13 Organizes cytoskeleton and
cell adhesion structures
88
Transforming protein RhoA H12 RHOA 3p21.3 Regulates cytoskeletal
dynamics
56, 88
RhoGDP dissociation inhibitor ARHGDIA 17q25.3 Regulates Rho GTPase
signaling
88
Pleiotrophin precuros; osteoblast specific factor 1 PTN/OSF-1 7q33 Stimulates mitogenic
growth of fibroblasts and
osteoblasts
56, 88
Amyloid A4 protein precursor; nexin II PN-II 21q21.3 Cell surface receptor 88
Defender against cell death 1 DAD1 14q11.2 Prevents apoptosis 88
Heat Shock 27-kDa protein (HSP27) HSP27 7q11.23 Actin organization and
translocation from
cytoplasm to nucleus upon
stress induction
56, 88
Macrophage-specific stimulating factor MCSF/CSF1 1p13.3 Controls the production,
differentiation and function
of macrophages
88
Transcription factor AP-1 AP1 1p32-p31 Key mediator of
macrophage education and
point of recruitment for
immunosuppressive
regulatory T cells
88, 105
human Early growth response protein 1 hEGR1 5q31.1 Promotes mitosis 88
Monocyte chemotactic protein 1 MCP1 17q11.2-q12 Chemotactic cytokine for
monocytes and basophils
88
Bone Proteoglycan II precusor; Decorin DCN 12q21.33 Matrix proteoglycan 56, 88
T-Cell specific rantes protein precursor RANTES 17q12 Chemoattractant for
monocytes, memory T cells
and eosinophils
88
Integrin Beta-1 ITGB1 10p11.2 Membrane receptor
involved in cell adhesion
and recognition in a variety
of processes including
immune response, tissue
repair and hemostasis
56
Osteonectin SPARC 5q31.3-q32 Matrix protein that
facilitates collagen
ossification
56
Ubiquitin RBX1 6q25.2-q27 Targets substrate proteins
for proteasomal degradation
56
Transcription factor ATF-4 ATF4 22q13.1 Transcriptional regulation
of osteoblasts and down-
regulates apelin to promote
apoptosis
56, 106
Elastase IIB ELA2B 1p36.21 Serine protease that
hydrolyzes matrix protein
56
c-myc MYC 8q24.21 Transcription factor that
regulates cell cycle
progression, apoptosis, and
cellular transformations
56
60 S ribosomal protein L13A RPL13A 19q13.3 Repression of inflammatory
genes
56
Prothymosin alpha PTMA 2q37.1 Influences chromatin
remodeling, anti-apoptotic
factor
56
Fibroblast tropomyosin TPM1 15q22.1 Actin-binding protein
involved in contractile
system of striated and
smooth muscle
56
Myosin light chain MYL2 12q24.11 Regulatory light chain
associated with myosin
Beta heavy chain
56
Filamin FLN Xq28 Actin-binding protein that
crosslinks actin filaments
and links actin to
membrane glycoproteins.
Interacts with integrins
56
Calcineurin A subunit alpha PPP3CA 4q24 Promotes cell migration
and invasion and inhibits
apoptosis
56 107
DNA binding protein inhibitor Id-2 ID2 2p25 Transcriptional regulator
that inhibits the function of
basic helix-loop-helix
transcription factors by
preventing their
heterodimerization,
negatively regulates cell
differentiation
56
Smooth muscle gamma actin ACTA2 10q23.3 Plays a role in cell motility,
structure and integrity
56
Desmin DES 2q35 Forms intracytoplasmic
filamentous network
connecting myofibrils
56
Cadherin FIB2 PCDHGB4 5q31 Cell adhesion proteins
expressed in fibroblasts and
playing a role in wound
healing
56
Cadherin FIB1 DCHS1 11p15.4 Cell adhesion proteins
expressed in fibroblasts and
playing a role in wound
healing
56
SMAD family member 7 SMAD7 18q21.1 Interacts with and promotes
degradation of TGFBR1
56
Insulin-like growth factor binding protein 6 IGFBP6 12q13 Negative regulator of
cellular senescence in
human fibroblasts
56, 108
Collagen 1 alpha COL1A1 17q21.33 Encodes pro-alpha 1 chains
of type 1 collagen
56
Transforming growth factor, beta 1 TGFB1 19q13.1 Cytokine that regulates
proliferation,
differentiation, adhesion
and cell migration
56

The monocyte chemotactic precursor protein 1 (MCP-1) gene, which not only recruits and activates peripheral blood monocytes but also drives the inflammatory cascade and promotes ossification, was the next most up-regulated gene.51 Higher levels of MCP-1 mRNA have been observed in PD plaque-derived fibroblasts when compared to fibroblasts derived from normal TA following incubation with recombinant TGFβ1.52 Lin et al. exposed three types of fibroblasts to TGFβ1 – PD plaque-derived fibroblasts, non-plaque derived fibroblasts from patients with PD, and fibroblasts from the TA of patients without PD – and subsequently measured MCP-1 expression.53 The highest levels of MCP-1 expression were found in PD plaque-derived fibroblasts using immunohistochemistry, followed by non-plaque derived fibroblasts from the same man, and then in fibroblasts from men without PD.

Among the six remaining highly up-regulated genes identified by Magee et al. using the Clontech platform, five are functionally associated with general cell proliferation, including the genes for the human early growth response protein 1 (hEGR1), the c-myc oncogene (MYC), 60S ribosomal protein L13A (RPL13A), transforming protein rhoAH1b (RHOA), and prothymosin alpha (PTMA).54, 55 The increased expression of these cellular proliferation genes, together with subsequent data from Mulhall and colleagues56 showing a blunted p53 gene activation response following irradiation of PD plaque-derived fibroblast cultures compared to normal TA fibroblast cultures, underscores the aberrant growth potential of PD cells resulting from loss of cell cycle control. Although no reports of malignant degeneration in PD plaques exist, PD plaque-derived fibroblasts transplanted into subcutaneous tissue of severe combined immunodeficient (SCID) mice resulted in the development of subcutaneous nodules containing multinucleated, pleomorphic cells with abundant cytoplasm and numerous mitotic figures.57 In contrast, subcutaneous placement of fibroblasts derived from men with congenital penile curvature or neonatal foreskin did not result in formation of such subcutaneous nodules.

Of the four most significantly down-regulated genes identified using the Clontech platform, the Inhibitor of DNA binding 2 (ID2) was the most down-regulated, followed by calcineurin A subunit alpha (PPP3CA), transcription factor ATF4 (ATF4), and ubiquitin. ID2 belongs to a family of genes that regulate cell lineage commitment and help maintain the balance between cell growth and apoptosis by inhibiting basic-helix-loop-helix transcription factors such as myoD, myogenin, MASH, Tal, and E2A, as well as cell cycle regulatory proteins, namely the retinoblastoma tumor suppressor protein (pRB).58 Levels of ID2 increase during cell proliferative states and decrease when cells reach a finite number of divisions and enter senescence.59 Although the role of ID2 has not been evaluated in fibrosis associated with PD, ID2’s role in cell fate determination and differentiation is well established in multiple tissue types including neural,62 hepatobiliary,65 and genitourinary.66, 67 Moreover, its overexpression attenuates liver and pulmonary fibrosis.68, 69

Using the Affymetrix platform, which analyzes the expression of 7129 genes compared to the 1176 genes represented in the Clontech platform,70 Magee et al.47 identified 14 up-regulated and 7 down-regulated genes in PD plaque tissue. Up-regulated genes were responsible primarily for the differentiation of fibroblasts into myofibroblasts (fibroblast muscle type tropomyosin (TPM), 20-kDa myosin light chain (MYL), filamin (FLN), gamma-smooth muscle actin (ACTA2), smooth muscle alpha actin (ACTA2), desmin (DES), 22-kDa smooth muscle protein), fibroblast attachment and collagen production (cadherin, TGFβ1, and insulin-like growth factor binding protein-6), elastin degradation (elastase IIB24), and the cellular stress response (28-kDa heat shock protein). Down-regulated genes included mothers against decapentaplegic homolog 7 (SMAD7), SPARC/osteonectin, decorin collagenase IV, and the HLA-B antigen.

Notable among the up- and down-regulated genes in this study were elastase IIB and SMAD7, which had the largest differential expressions between PD and normal tissue. The overexpression of elastase may play a role in cell proliferation through the cleavage of insulin receptor substrate-1 (IRS1) and downstream activation of the phosphatidylinositol-3 kinase (PI3K) pathway.71 Elastase may also promote cell proliferation via release of growth factors such as transforming growth factor-α (TGFα), platelet derived growth factor (PDGF) and vascular endothelial growth factor (VEGF).72 Decreased expression of SMAD7 is likely to have a pro-fibrotic role in PD, given that SMADs are involved in TGFβ signaling.73 SMAD7 expression is normally increased following TGFβ1-mediated signaling, and SMAD7 antagonizes TGFβ1 signaling in multiple ways including dephosphorylation and resulting inactivation of the type 1 TGF β 1 receptor (TGFB1R), recruitment of SMAD2/3 and the ubiquitination and eventual degradation of the TGFβ1 type 1 receptor.7476 Although subsequent work by Szardening-Kirchner et al.52 did not observe decreased SMAD7 expression following incubation of PD fibroblasts with recombinant TGFβ1, gene expression levels of SMAD2-4 did not change despite fibroblast stimulation with TGFβ1, suggesting potential dysfunction of SMAD7-mediated recruitment of SMAD2/3. The potential under-expression or loss of function of SMAD7 may therefore remove an important checkpoint in the inflammatory cascade, predisposing to PD.

In contrast, overexpression of SMAD7 can limit the fibrotic response of PD fibroblasts to TGFβ1. Choi et al.77 recently demonstrated the impact of SMAD7 overexpression in PD by isolating and transfecting fibroblasts from human PD plaques with either a vector containing the full-length human SMAD7 gene or an empty vector control. Fibroblasts transfected with SMAD7 demonstrated profound decreases in extracellular matrix protein expression, including decreases in plasminogen activating inhibitor-1 (PAI1), fibronectin, collagen I, and collagen IV when compared with PD plaque-derived fibroblasts transfected with empty vector. SMAD7 was also shown to have pro-apoptotic effects on both TGFβ1-stimulated and unstimulated PD plaque-derived fibroblasts through the decreased expression of cyclin D1.

In order to better understand the dysregulation of apoptosis in the PD plaque, Zorba et al.78 recently compared gene expression in PD plaque tissue to that of adjacent grossly normal TA in 8 men undergoing plaque incision and grafting procedures. RNA expression levels were quantified for six pro-apoptotic genes – Fas, Fas Ligand, Bcl-2, p53, Caspase 3 and Caspase 8 – using reverse transcriptase PCR (RT PCR). There was no Fas expression and Fas Ligand had lower expression in PD plaque tissue compared with normal TA. In contrast, higher relative expression levels of Bcl-2, p53, Caspase 3 and Caspase 8 were observed in PD plaque tissue. However, as previously demonstrated by Mulhall et al.30, PD may represent a TA field defect, and validation studies comparing gene expression of pro-apoptotic genes in PD plaque to normal-appearing TA derived from men without PD are needed.

Differences in Gene Expression Between PD and DD Lesions

Array-based gene expression analysis has been used to compare gene expression between PD and DD lesions, and genes with altered expression that overlap between the conditions have been identified. Qian et al.79 compared the gene expression profiles of 9 patients with PD and 9 patients with DD to matched controls. Genes with at least a 2-fold relative difference in expression were considered significantly affected. The authors identified 15 genes with overexpression in the PD plaque, of which 8 were functionally related to fibrosis in tissue from both PD and DD lesions and included matrix metalloproteinase 2 (MMP2), MMP9, Thymosin Beta-10 (TMSB10), TMSB4, Cortactin/amplaxin/ems-1 oncogene (CTTN), Transforming protein RhoA H12 (RHOA), RhoGDP dissociation inhibitor 1 (ARHGDIA), and PTN/OSF-1. Among these eight genes, MMP2 and MMP9 were up-regulated in the PD plaque but not in the isolated PD fibroblasts.

The gene with the highest expression in PD plaque tissue in this study was MMP9. MMP9 belongs to the matrix metalloproteinase family of endopeptidases which catabolize gelatins, collagen type I, and collagen type III.80 The expression of MMPs is stimulated by inflammatory cytokines including IL1 and TNFα, and regulated by the co-expression of tissue inhibitors of metalloproteinases (TIMPs). In situ activation of MMPs depends on thymosins, which were elevated in not only PD plaque but also PD plaque-derived fibroblasts isolated from the PD plaque and DD nodules.81 Del Carlo et al. characterized the expression of MMPs and TIMPs using MMP protein microarrays and Western blot analysis of PD fibroblasts following transcriptional induction using either IL1β or TGFβ. IL1β significantly increased the production of MMP1, 3, 10, and 13 and only mildly increased the production of MMP8 and MMP9.82 Although TGFβ did not up-regulate MMP1, 8 or 13 and only mildly increased MMP9 levels, it resulted in significant up-regulation of TIMP 1–4. The authors also evaluated the relative expression of MMP1, 8, and 13 as well as TIMP 1–4 using Western blot analysis of plaque tissue samples obtained from 13 men with stable PD lesions compared with normal TA tissue from 6 men without PD. MMP1, 8 and 13 were detected in both PD and non-PD TA tissue. However, TIMPs 1–4 were readily detectable in TA from men with PD. These findings underscore the role of both MMPs and TIMPs in the fibrotic process of PD. MMPs may contribute to fibrosis through the altered function of extracellular matrix proteins following proteolysis or release of sequestered growth factors such as TGFα, PDGF, and VEGF, within the extracellular matrix.72, 83 The local accumulation of TIMPs also promotes fibrosis by decreasing the net catabolic activity of MMPs and allowing the gradual buildup of collagen and other matrix proteins.82

Other over-expressed genes identified in either PD plaque tissue and/or PD plaque-derived fibroblasts that merit inclusion here include transcription factor AP1, hEGR1, MCP1, bone proteoglycan II precursor (PGS2)/decorin (DCN), T-Cell specific RANTES protein precursor, and integrin beta 1 (ITGB1).79 AP1 plays a central role in the inflammatory cascade by regulating the transcription of cytokines, which influence immune cell proliferation, differentiation, migration, and apoptosis.84, 85 The second and third most over-expressed genes included hEGR1 and MCP1, whose roles in the development of PD were discussed above.

Epigenetic Regulation

Epigenetic regulation refers to heritable alterations in gene expression that occur without altering the gene sequence, but rather dynamically alter the transcriptional potential of a cell. This type of gene regulation can occur at the transcriptional or post-transcriptional level through modifications of histone proteins, such as acetylation, methylation, and phosphorylation, or through noncoding RNA-based regulation that can modify chromatin structure. Histone deacetylases (HDACs) are enzymes that control the balance of histone acetylation/deacetylation and alter the transcriptional potential of a given gene by regulating access of its promoter to transcriptional regulatory proteins. HDACs selectively remove acetyl groups from the terminal lysine on histones, causing the DNA to tightly wrap around the histone protein, and limiting access to transcriptional machinery.90 Importantly, HDACs have been implicated in the pathogenesis of multiple fibrotic disorders of the kidneys, bladder, lungs, heart and liver.9193

Inhibition of HDACs can repress TGFβ-mediated signaling. In order to determine if inhibition of HDAC2 would limit the profibrotic response to TGFβ signaling in human PD plaques, Ryu et al. used RNA interference (RNAi) via small interfering RNA (siRNA) targeting HDAC2, comparing PD plaque-derived fibroblasts to control fibroblasts transfected with nonspecific scramble siRNA.94 Protein expression levels of plasminogen activator inhibitor 1 (PAI1), fibronectin, collagens I and IV, smooth muscle α-actin, and HDAC2 were assessed using Western blot following stimulation with TGFβ. The authors observed a 60% reduction in HDAC2 expression in PD plaque-derived fibroblasts transfected with the HDAC2-targeted siRNA, with similar reductions in expression of PAI1, fibronectin, collagens I and IV, and smooth muscle α-actin. Additionally, the conversion of fibroblasts into myofibroblasts, the causal cell type in aberrant fibrosis, was blocked in fibroblasts treated with HDAC2-targeted siRNA. Reduced phosphorylation and nuclear migration of SMAD2/3, which typically occurs following TGFβ-mediated stimulation of PD plaque fibroblasts, were also observed.

Kwon et al. corroborated these findings using adenovirus-encoded HDAC2 small hairpin RNA (ad-HDAC2 shRNA) in a rat model.95 The authors observed that PD model rats receiving injections of Ad-HDAC2 shRNA had regression of fibrotic penile plaque, fewer intralesional inflammatory cells, impaired nuclear translocation of phosphorylated Smad3, inhibited differentiation of fibroblasts into myofibroblasts, and reduced collagen production. Furthermore, cultured rat fibroblasts from PD plaques exhibited reduced expression of cyclin D1, which blocked cell cycle entry and a propensity towards apoptosis in both basal and TGFβ-stimulated conditions.

Conclusions

The genetic underpinnings of fibrotic diatheses, including Peyronie’s and Dupuytren’s diseases, are just beginning to be understood. From studies 3 decades ago that established genetic links to these conditions, much progress has been made in defining the similarities between Dupuytren’s and Peyronie’s diseases, particularly on a cellular levels. However, the specific genetic factors that predispose patients to PD and/or DD are not currently known, nor are the relationships between PD/DD and other heritable conditions, including malignancy. While translation of our current understanding of the genetic factors predisposing to PD to clinical practice is in its infancy, future research will provide a more comprehensive understanding of the landscape of genetic factors responsible for the development of PD, and will facilitate development of novel biomarkers and therapies to allow earlier detection and treatment of those afflicted with PD. As such, the present represents an exciting time for elucidation of the genetic landscape of these conditions, towards the goal of an improved understanding of these related fibrotic diatheses, as well as an improved ability to risk stratify affected patients, and ultimately provide better care for them through more effective targeted therapies and disease prevention.

Acknowledgments

Funding: A.W.P. and A.S.H. are National Institutes of Health (NIH) K12 Scholars supported by a Male Reproductive Health Research Career (MHRH) Development Physician-Scientist Award (HD073917-01) from the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Program (to Dolores J. Lamb).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Pryor JP, Ralph DJ. Clinical presentations of Peyronie's disease. International journal of impotence research. 2002;14:414–417. doi: 10.1038/sj.ijir.3900877. [DOI] [PubMed] [Google Scholar]
  • 2.Devine CJ, Jr, Somers KD, Jordan SG, Schlossberg SM. Proposal: trauma as the cause of the Peyronie's lesion. The Journal of urology. 1997;157:285–290. doi: 10.1016/s0022-5347(01)65361-8. [DOI] [PubMed] [Google Scholar]
  • 3.Brock G, Hsu GL, Nunes L, von Heyden B, Lue TF. The anatomy of the tunica albuginea in the normal penis and Peyronie's disease. The Journal of urology. 1997;157:276–281. [PubMed] [Google Scholar]
  • 4.Somers KD, Dawson DM. Fibrin deposition in Peyronie's disease plaque. The Journal of urology. 1997;157:311–315. [PubMed] [Google Scholar]
  • 5.Somers KD, Sismour EN, Wright GL, Jr, Devine CJ, Jr, Gilbert DA, Horton CE. Isolation and characterization of collagen in Peyronie's disease. The Journal of urology. 1989;141:629–631. doi: 10.1016/s0022-5347(17)40920-7. [DOI] [PubMed] [Google Scholar]
  • 6.Moreland RB, Nehra A. Pathophysiology of Peyronie's disease. International journal of impotence research. 2002;14:406–410. doi: 10.1038/sj.ijir.3900875. [DOI] [PubMed] [Google Scholar]
  • 7.Nugteren HM, Nijman JM, de Jong IJ, van Driel MF. The association between Peyronie's and Dupuytren's disease. International journal of impotence research. 2011;23:142–145. doi: 10.1038/ijir.2011.18. [DOI] [PubMed] [Google Scholar]
  • 8.Michou L, Lermusiaux JL, Teyssedou JP, Bardin T, Beaudreuil J, Petit-Teixeira E. Genetics of Dupuytren's disease. Joint Bone Spine. 2012;79:7–12. doi: 10.1016/j.jbspin.2011.05.027. [DOI] [PubMed] [Google Scholar]
  • 9.Noss MB, Day NS, Christ GJ, Melman A. The genetics and immunology of Peyronie's disease. Int J Impot Res. 2000;12(Suppl 4):S127–S132. doi: 10.1038/sj.ijir.3900591. [DOI] [PubMed] [Google Scholar]
  • 10.Haag SM, Hauck EW, Eickelberg O, Szardening-Kirchner C, Diemer T, Weidner W. Investigation of the antifibrotic effect of IFN-gamma on fibroblasts in a cell culture model of Peyronie's disease. European urology. 2008;53:425–430. doi: 10.1016/j.eururo.2007.06.036. [DOI] [PubMed] [Google Scholar]
  • 11.El-Sakka AI, Hassoba HM, Pillarisetty RJ, Dahiya R, Lue TF. Peyronie's disease is associated with an increase in transforming growth factor-beta protein expression. The Journal of urology. 1997;158:1391–1394. [PubMed] [Google Scholar]
  • 12.Bias WB, Nyberg LM, Jr, Hochberg MC, Walsh PC. Peyronie's disease: a newly recognized autosomal-dominant trait. American journal of medical genetics. 1982;12:227–235. doi: 10.1002/ajmg.1320120213. [DOI] [PubMed] [Google Scholar]
  • 13.Nyberg LM, Jr, Bias WB, Hochberg MC, Walsh PC. Identification of an inherited form of Peyronie's disease with autosomal dominant inheritance and association with Dupuytren's contracture and histocompatibility B7 cross-reacting antigens. The Journal of urology. 1982;128:48–51. doi: 10.1016/s0022-5347(17)52751-2. [DOI] [PubMed] [Google Scholar]
  • 14.Shaw RB, Jr, Chong AK, Zhang A, Hentz VR, Chang J. Dupuytren's disease: history, diagnosis, and treatment. Plast Reconstr Surg. 2007;120:44e–54e. doi: 10.1097/01.prs.0000278455.63546.03. [DOI] [PubMed] [Google Scholar]
  • 15.Fitzgerald AM, Kirkpatrick JJ, Naylor IL. Dupuytren's disease The way forward? J Hand Surg Br. 1999;24:395–399. doi: 10.1054/jhsb.1999.0207. [DOI] [PubMed] [Google Scholar]
  • 16.Willscher MK, Cwazka WF, Novicki DE. The association of histocompatibility antigens of the B7 cross-reacting group with Peyronie's disease. The Journal of urology. 1979;122:34–35. doi: 10.1016/s0022-5347(17)56238-2. [DOI] [PubMed] [Google Scholar]
  • 17.Sommer F, Schwarzer U, Wassmer G, Bloch W, Braun M, Klotz T, Engelmann U. Epidemiology of Peyronie's disease. International journal of impotence research. 2002;14:379–383. doi: 10.1038/sj.ijir.3900863. [DOI] [PubMed] [Google Scholar]
  • 18.Lindsay MB, Schain DM, Grambsch P, Benson RC, Beard CM, Kurland LT. The incidence of Peyronie's disease in Rochester, Minnesota, 1950 through 1984. The Journal of urology. 1991;146:1007–1009. doi: 10.1016/s0022-5347(17)37988-0. [DOI] [PubMed] [Google Scholar]
  • 19.Tal R, Hall MS, Alex B, Choi J, Mulhall JP. Peyronie's disease in teenagers. The journal of sexual medicine. 2012;9:302–308. doi: 10.1111/j.1743-6109.2011.02502.x. [DOI] [PubMed] [Google Scholar]
  • 20.Tefekli A, Kandirali E, Erol H, Alp T, Koksal T, Kadioglu A. Peyronie's disease in men under age 40: characteristics and outcome. International journal of impotence research. 2001;13:18–23. doi: 10.1038/sj.ijir.3900635. [DOI] [PubMed] [Google Scholar]
  • 21.Schwarzer U, Sommer F, Klotz T, Braun M, Reifenrath B, Engelmann U. The prevalence of Peyronie's disease: results of a large survey. BJU international. 2001;88:727–730. doi: 10.1046/j.1464-4096.2001.02436.x. [DOI] [PubMed] [Google Scholar]
  • 22.Perimenis P, Athanasopoulos A, Gyftopoulos K, Katsenis G, Barbalias G. Peyronie's disease: epidemiology and clinical presentation of 134 cases. International urology and nephrology. 2001;32:691–694. doi: 10.1023/a:1014485204205. [DOI] [PubMed] [Google Scholar]
  • 23.Bjekic MD, Vlajinac HD, Sipetic SB, Marinkovic JM. Risk factors for Peyronie's disease: a case-control study. BJU Int. 2006;97:570–574. doi: 10.1111/j.1464-410X.2006.05969.x. [DOI] [PubMed] [Google Scholar]
  • 24.Ventimiglia E, Capogrosso P, Colicchia M, Boeri L, Serino A, La Croce G, Russo A, Capitanio U, Briganti A, Cantiello F, Mirone V, Damiano R, Montorsi F, Salonia A. Peyronie's Disease and Autoimmunity-A Real-Life Clinical Study and Comprehensive Review. J Sex Med. 2015 doi: 10.1111/jsm.12825. [DOI] [PubMed] [Google Scholar]
  • 25.Leffell MS, Devine CJ, Jr, Horton CE, Somers KD, Dawson D, Vande Berg JS, Bluemink GG, Wright GI., Jr Non-association of Peyronie's disease with HLA B7 cross-reactive antigens. The Journal of urology. 1982;127:1223–1224. doi: 10.1016/s0022-5347(17)54302-5. [DOI] [PubMed] [Google Scholar]
  • 26.Deguchi T, Maeda S, Sakai S, Kuriyama M, Kawada Y, Nishiura T. HLA-A and B antigens in patients with Peyronie disease. Urology. 1984;23:547–548. doi: 10.1016/0090-4295(84)90068-2. [DOI] [PubMed] [Google Scholar]
  • 27.Hauck EW, Hauptmann A, Weidner W, Bein G, Hackstein H. Prospective analysis of HLA classes I and II antigen frequency in patients with Peyronie's disease. The Journal of urology. 2003;170:1443–1446. doi: 10.1097/01.ju.0000076488.89748.e1. [DOI] [PubMed] [Google Scholar]
  • 28.Somers KD, Winters BA, Dawson DM, Leffell MS, Wright GL, Jr, Devine CJ, Jr, Gilbert DA, Horton CE. Chromosome abnormalities in Peyronie's disease. The Journal of urology. 1987;137:672–675. doi: 10.1016/s0022-5347(17)44170-x. [DOI] [PubMed] [Google Scholar]
  • 29.Guerneri S, Stioui S, Mantovani F, Austoni E, Simoni G. Multiple clonal chromosome abnormalities in Peyronie's disease. Cancer Genet Cytogenet. 1991;52:181–185. doi: 10.1016/0165-4608(91)90461-3. [DOI] [PubMed] [Google Scholar]
  • 30.Mulhall JP, Nicholson B, Pierpaoli S, Lubrano T, Shankey TV. Chromosomal instability is demonstrated by fibroblasts derived from the tunica of men with Peyronie's disease. International journal of impotence research. 2004;16:288–293. doi: 10.1038/sj.ijir.3901170. [DOI] [PubMed] [Google Scholar]
  • 31.Raghu G, Masta S, Meyers D, Narayanan AS. Collagen synthesis by normal and fibrotic human lung fibroblasts and the effect of transforming growth factor-beta. Am Rev Respir Dis. 1989;140:95–100. doi: 10.1164/ajrccm/140.1.95. [DOI] [PubMed] [Google Scholar]
  • 32.Milani S, Herbst H, Schuppan D, Stein H, Surrenti C. Transforming growth factors beta 1 and beta 2 are differentially expressed in fibrotic liver disease. Am J Pathol. 1991;139:1221–1229. [PMC free article] [PubMed] [Google Scholar]
  • 33.Domes T, De Young L, O'Gorman DB, Gan BS, Bella AJ, Brock G. Is there a role for proteomics in Peyronie's disease? The journal of sexual medicine. 2007;4:867–877. doi: 10.1111/j.1743-6109.2007.00470.x. [DOI] [PubMed] [Google Scholar]
  • 34.El-Sakka AI, Hassoba HM, Chui RM, Bhatnagar RS, Dahiya R, Lue TF. An animal model of Peyronie's-like condition associated with an increase of transforming growth factor beta mRNA and protein expression. The Journal of urology. 1997;158:2284–2290. doi: 10.1016/s0022-5347(01)68236-3. [DOI] [PubMed] [Google Scholar]
  • 35.Piao S, Ryu JK, Shin HY, Zhang L, Song SU, Han JY, Park SH, Kim JM, Kim IH, Kim SJ, Suh JK. Repeated intratunical injection of adenovirus expressing transforming growth factor-beta1 in a rat induces penile curvature with tunical fibrotic plaque: a useful model for the study of Peyronie's disease. Int J Androl. 2008;31:346–353. doi: 10.1111/j.1365-2605.2007.00780.x. [DOI] [PubMed] [Google Scholar]
  • 36.Ryu JK, Piao S, Shin HY, Choi MJ, Zhang LW, Jin HR, Kim WJ, Han JY, Hong SS, Park SH, Lee SJ, Kim IH, Lee CR, Kim DK, Mamura M, Kim SJ, Suh JK. IN-1130, a novel transforming growth factor-beta type I receptor kinase (activin receptor-like kinase 5) inhibitor, promotes regression of fibrotic plaque and corrects penile curvature in a rat model of Peyronie's disease. The journal of sexual medicine. 2009;6:1284–1296. doi: 10.1111/j.1743-6109.2009.01216.x. [DOI] [PubMed] [Google Scholar]
  • 37.Choi ME. Mechanism of transforming growth factor-beta1 signaling. Kidney Int Suppl. 2000;77:S53–S58. [PubMed] [Google Scholar]
  • 38.Yuan X, Wei Q, Komaki R, Liu Z, Yang J, Tucker SL, Xu T, Heymach JV, Lu C, Cox JD, Liao Z. Polymorphisms Predict Distant Metastasis-Free Survival in Patients with Inoperable Non-Small-Cell Lung Cancer after Definitive Radiotherapy. PLoS One. 2013;8:e65659. doi: 10.1371/journal.pone.0065659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dunning AM, Ellis PD, McBride S, Kirschenlohr HL, Healey CS, Kemp PR, Luben RN, Chang-Claude J, Mannermaa A, Kataja V, Pharoah PD, Easton DF, Ponder BA, Metcalfe JC. A transforming growth factorbeta1 signal peptide variant increases secretion in vitro and is associated with increased incidence of invasive breast cancer. Cancer Res. 2003;63:2610–2615. [PubMed] [Google Scholar]
  • 40.Jaakkola E, Crane AM, Laiho K, Herzberg I, Sims AM, Bradbury L, Calin A, Brophy S, Kauppi M, Kaarela K, Wordsworth BP, Tuomilehto J, Brown MA. The effect of transforming growth factor beta1 gene polymorphisms in ankylosing spondylitis. Rheumatology (Oxford) 2004;43:32–38. doi: 10.1093/rheumatology/keg457. [DOI] [PubMed] [Google Scholar]
  • 41.Dickson MR, Perry RT, Wiener H, Go RC. Association studies of transforming growth factor-beta 1 and Alzheimer's disease. Am J Med Genet B Neuropsychiatr Genet. 2005;139B:38–41. doi: 10.1002/ajmg.b.30218. [DOI] [PubMed] [Google Scholar]
  • 42.Awad MR, El-Gamel A, Hasleton P, Turner DM, Sinnott PJ, Hutchinson IV. Genotypic variation in the transforming growth factor-beta1 gene: association with transforming growth factor-beta1 production, fibrotic lung disease, and graft fibrosis after lung transplantation. Transplantation. 1998;66:1014–1020. doi: 10.1097/00007890-199810270-00009. [DOI] [PubMed] [Google Scholar]
  • 43.Hauck EW, Hauptmann A, Schmelz HU, Bein G, Weidner W, Hackstein H. Prospective analysis of single nucleotide polymorphisms of the transforming growth factor beta-1 gene in Peyronie's disease. The Journal of urology. 2003;169:369–372. doi: 10.1016/S0022-5347(05)64129-8. [DOI] [PubMed] [Google Scholar]
  • 44.Dolmans GH, Werker PM, Hennies HC, Furniss D, Festen EA, Franke L, Becker K, van der Vlies P, Wolffenbuttel BH, Tinschert S, Toliat MR, Nothnagel M, Franke A, Klopp N, Wichmann HE, Nurnberg P, Giele H, Ophoff RA, Wijmenga C, Dutch Dupuytren Study G, German Dupuytren Study G, LifeLines Cohort S, Consortium B-G. Wnt signaling and Dupuytren's disease. The New England journal of medicine. 2011;365:307–317. doi: 10.1056/NEJMoa1101029. [DOI] [PubMed] [Google Scholar]
  • 45.Dolmans GH, Werker PM, de Jong IJ, Nijman RJ, LifeLines Cohort S, Wijmenga C, Ophoff RA. WNT2 locus is involved in genetic susceptibility of Peyronie's disease. The journal of sexual medicine. 2012;9:1430–1434. doi: 10.1111/j.1743-6109.2012.02704.x. [DOI] [PubMed] [Google Scholar]
  • 46.Panteris E, Swift S, Payne A, Liu X. Mining pathway signatures from microarray data and relevant biological knowledge. J Biomed Inform. 2007;40:698–706. doi: 10.1016/j.jbi.2007.01.004. [DOI] [PubMed] [Google Scholar]
  • 47.Magee TR, Qian A, Rajfer J, Sander FC, Levine LA, Gonzalez-Cadavid NF. Gene expression profiles in the Peyronie's disease plaque. Urology. 2002;59:451–457. doi: 10.1016/s0090-4295(01)01578-3. [DOI] [PubMed] [Google Scholar]
  • 48.Imai S, Kaksonen M, Raulo E, Kinnunen T, Fages C, Meng X, Lakso M, Rauvala H. Osteoblast recruitment and bone formation enhanced by cell matrix-associated heparin-binding growth-associated molecule (HB-GAM) J Cell Biol. 1998;143:1113–1128. doi: 10.1083/jcb.143.4.1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bouderlique T, Henault E, Lebouvier A, Frescaline G, Bierling P, Rouard H, Courty J, Albanese P, Chevallier N. Pleiotrophin commits human bone marrow mesenchymal stromal cells towards hypertrophy during chondrogenesis. PLoS One. 2014;9:e88287. doi: 10.1371/journal.pone.0088287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tare RS, Oreffo RO, Clarke NM, Roach HI. Pleiotrophin/Osteoblast-stimulating factor 1: dissecting its diverse functions in bone formation. J Bone Miner Res. 2002;17:2009–2020. doi: 10.1359/jbmr.2002.17.11.2009. [DOI] [PubMed] [Google Scholar]
  • 51.Yoshimura H, Nakahama K, Safronova O, Tanaka N, Muneta T, Morita I. Transforming growth factor-beta stimulates IL-1beta-induced monocyte chemoattractant protein-1 expression in human synovial cells via the ERK/AP-1 pathway. Inflamm Res. 2006;55:543–549. doi: 10.1007/s00011-006-5144-9. [DOI] [PubMed] [Google Scholar]
  • 52.Szardening-Kirchner C, Konrad L, Hauck EW, Haag SM, Eickelberg O, Weidner W. Upregulation of mRNA expression of MCP-1 by TGF-beta1 in fibroblast cells from Peyronie's disease. World J Urol. 2009;27:123–130. doi: 10.1007/s00345-008-0320-x. [DOI] [PubMed] [Google Scholar]
  • 53.Lin CS, Lin G, Wang Z, Maddah SA, Lue TF. Upregulation of monocyte chemoattractant protein 1 and effects of transforming growth factor-beta 1 in Peyronie's disease. Biochem Biophys Res Commun. 2002;295:1014–1019. doi: 10.1016/s0006-291x(02)00765-9. [DOI] [PubMed] [Google Scholar]
  • 54.Pucci B, Kasten M, Giordano A. Cell cycle and apoptosis. Neoplasia. 2000;2:291–299. doi: 10.1038/sj.neo.7900101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chen FW, Ioannou YA. Ribosomal proteins in cell proliferation and apoptosis. Int Rev Immunol. 1999;18:429–448. doi: 10.3109/08830189909088492. [DOI] [PubMed] [Google Scholar]
  • 56.Mulhall JP, Branch J, Lubrano T, Shankey TV. Perturbation of cell cycle regulators in Peyronie's disease. International journal of impotence research. 2001;13(Suppl 5):S21–S28. doi: 10.1038/sj.ijir.3900771. [DOI] [PubMed] [Google Scholar]
  • 57.Mulhall JP, Martin DJ, Lubrano T, Moser M, Kwon E, Wojcik E, Shankey TV. Peyronie's disease fibroblasts demonstrate tumorigenicity in the severe combined immunodeficient (SCID) mouse model. International journal of impotence research. 2004;16:99–104. doi: 10.1038/sj.ijir.3901183. [DOI] [PubMed] [Google Scholar]
  • 58.Zebedee Z, Hara E. Id proteins in cell cycle control and cellular senescence. Oncogene. 2001;20:8317–8325. doi: 10.1038/sj.onc.1205092. [DOI] [PubMed] [Google Scholar]
  • 59.Hara E, Yamaguchi T, Nojima H, Ide T, Campisi J, Okayama H, Oda K. Id-related genes encoding helix-loop-helix proteins are required for G1 progression and are repressed in senescent human fibroblasts. J Biol Chem. 1994;269:2139–2145. [PubMed] [Google Scholar]
  • 60.Olson EN, Klein WH. bHLH factors in muscle development: dead lines and commitments, what to leave in and what to leave out. Genes Dev. 1994;8:1–8. doi: 10.1101/gad.8.1.1. [DOI] [PubMed] [Google Scholar]
  • 61.Yang L, Ma X, Lyone A, Zou J, Blackburn ML, Pan J, Yang D, Matsushita H, Mei B, Zielinska-Kwiatkowska A, Chansky HA. Proper expression of helix-loop-helix protein Id2 is important to chondrogenic differentiation of ATDC5 cells. Biochem J. 2009;419:635–643. doi: 10.1042/BJ20081715. [DOI] [PubMed] [Google Scholar]
  • 62.Lee JE. Basic helix-loop-helix genes in neural development. Curr Opin Neurobiol. 1997;7:13–20. doi: 10.1016/s0959-4388(97)80115-8. [DOI] [PubMed] [Google Scholar]
  • 63.Yokota Y, Mansouri A, Mori S, Sugawara S, Adachi S, Nishikawa S, Gruss P. Development of peripheral lymphoid organs and natural killer cells depends on the helix-loop-helix inhibitor Id2. Nature. 1999;397:702–706. doi: 10.1038/17812. [DOI] [PubMed] [Google Scholar]
  • 64.Masson F, Minnich M, Olshansky M, Bilic I, Mount AM, Kallies A, Speed TP, Busslinger M, Nutt SL, Belz GT. Id2-mediated inhibition of E2A represses memory CD8+ T cell differentiation. Journal of immunology. 2013;190:4585–4594. doi: 10.4049/jimmunol.1300099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rodriguez JL, Sandoval J, Serviddio G, Sastre J, Morante M, Perrelli MG, Martinez-Chantar ML, Vina J, Vina JR, Mato JM, Avila MA, Franco L, Lopez-Rodas G, Torres L. Id2 leaves the chromatin of the E2F4-p130-controlled c-myc promoter during hepatocyte priming for liver regeneration. Biochem J. 2006;398:431–437. doi: 10.1042/BJ20060380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chaudhary J, Schmidt M, Sadler-Riggleman I. Negative acting HLH proteins Id 1, Id 2, Id 3, and Id 4 are expressed in prostate epithelial cells. Prostate. 2005;64:253–264. doi: 10.1002/pros.20238. [DOI] [PubMed] [Google Scholar]
  • 67.Chaudhary J, Sadler-Riggleman I, Ague JM, Skinner MK. The helix-loop-helix inhibitor of differentiation (ID) proteins induce post-mitotic terminally differentiated Sertoli cells to re-enter the cell cycle and proliferate. Biol Reprod. 2005;72:1205–1217. doi: 10.1095/biolreprod.104.035717. [DOI] [PubMed] [Google Scholar]
  • 68.Yang J, Velikoff M, Agarwal M, Disayabutr S, Wolters PJ, Kim KK. Overexpression of inhibitor of DNA-binding 2 attenuates pulmonary fibrosis through regulation of c-Abl and Twist. Am J Pathol. 2015;185:1001–1011. doi: 10.1016/j.ajpath.2014.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kinoshita K, Iimuro Y, Otogawa K, Saika S, Inagaki Y, Nakajima Y, Kawada N, Fujimoto J, Friedman SL, Ikeda K. Adenovirus-mediated expression of BMP-7 suppresses the development of liver fibrosis in rats. Gut. 2007;56:706–714. doi: 10.1136/gut.2006.092460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chee M, Yang R, Hubbell E, Berno A, Huang XC, Stern D, Winkler J, Lockhart DJ, Morris MS, Fodor SP. Accessing genetic information with high-density DNA arrays. Science. 1996;274:610–614. doi: 10.1126/science.274.5287.610. [DOI] [PubMed] [Google Scholar]
  • 71.Houghton AM, Rzymkiewicz DM, Ji H, Gregory AD, Egea EE, Metz HE, Stolz DB, Land SR, Marconcini LA, Kliment CR, Jenkins KM, Beaulieu KA, Mouded M, Frank SJ, Wong KK, Shapiro SD. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat Med. 2010;16:219–223. doi: 10.1038/nm.2084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wada Y, Yoshida K, Tsutani Y, Shigematsu H, Oeda M, Sanada Y, Suzuki T, Mizuiri H, Hamai Y, Tanabe K, Ukon K, Hihara J. Neutrophil elastase induces cell proliferation and migration by the release of TGF-alpha, PDGF and VEGF in esophageal cell lines. Oncol Rep. 2007;17:161–167. [PubMed] [Google Scholar]
  • 73.Liu T, Feng XH. Regulation of TGF-beta signalling by protein phosphatases. Biochem J. 2010;430:191–198. doi: 10.1042/BJ20100427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Mori Y, Chen SJ, Varga J. Modulation of endogenous Smad expression in normal skin fibroblasts by transforming growth factor-beta. Exp Cell Res. 2000;258:374–383. doi: 10.1006/excr.2000.4930. [DOI] [PubMed] [Google Scholar]
  • 75.Nakao A, Afrakhte M, Moren A, Nakayama T, Christian JL, Heuchel R, Itoh S, Kawabata M, Heldin NE, Heldin CH, ten Dijke P. Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature. 1997;389:631–635. doi: 10.1038/39369. [DOI] [PubMed] [Google Scholar]
  • 76.Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H, Thomsen GH, Wrana JL. Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF beta receptor for degradation. Mol Cell. 2000;6:1365–1375. doi: 10.1016/s1097-2765(00)00134-9. [DOI] [PubMed] [Google Scholar]
  • 77.Choi MJ, Song KM, Park JM, Kwon MH, Kwon KD, Park SH, Ryu DS, Ryu JK, Suh JK. Effect of gene overexpression on TGF-beta1-induced profibrotic responses in fibroblasts derived from Peyronie's plaque. Asian J Androl. 2014 doi: 10.4103/1008-682X.142130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zorba OU, Sirma S, Ozgon G, Salabas E, Ozbek U, Kadioglu A. Comparison of apoptotic gene expression profiles between Peyronie's disease plaque and tunica albuginea. Adv Clin Exp Med. 2012;21:607–614. [PubMed] [Google Scholar]
  • 79.Qian A, Meals RA, Rajfer J, Gonzalez-Cadavid NF. Comparison of gene expression profiles between Peyronie's disease and Dupuytren's contracture. Urology. 2004;64:399–404. doi: 10.1016/j.urology.2004.04.006. [DOI] [PubMed] [Google Scholar]
  • 80.Nagase H, Woessner JF., Jr Matrix metalloproteinases. J Biol Chem. 1999;274:21491–21494. doi: 10.1074/jbc.274.31.21491. [DOI] [PubMed] [Google Scholar]
  • 81.Huff T, Muller CS, Otto AM, Netzker R, Hannappel E. beta-Thymosins, small acidic peptides with multiple functions. Int J Biochem Cell Biol. 2001;33:205–220. doi: 10.1016/s1357-2725(00)00087-x. [DOI] [PubMed] [Google Scholar]
  • 82.Del Carlo M, Cole AA, Levine LA. Differential calcium independent regulation of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases by interleukin-1beta and transforming growth factor-beta in Peyronie's plaque fibroblasts. The Journal of urology. 2008;179:2447–2455. doi: 10.1016/j.juro.2008.01.093. [DOI] [PubMed] [Google Scholar]
  • 83.Zuo J, Ferguson TA, Hernandez YJ, Stetler-Stevenson WG, Muir D. Neuronal matrix metalloproteinase-2 degrades and inactivates a neurite-inhibiting chondroitin sulfate proteoglycan. J Neurosci. 1998;18:5203–5211. doi: 10.1523/JNEUROSCI.18-14-05203.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Shaulian E. AP-1--The Jun proteins: Oncogenes or tumor suppressors in disguise? Cell Signal. 2010;22:894–899. doi: 10.1016/j.cellsig.2009.12.008. [DOI] [PubMed] [Google Scholar]
  • 85.Wagner EF, Eferl R. Fos/AP-1 proteins in bone and the immune system. Immunol Rev. 2005;208:126–140. doi: 10.1111/j.0105-2896.2005.00332.x. [DOI] [PubMed] [Google Scholar]
  • 86.de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J. 2003;370:737–749. doi: 10.1042/BJ20021321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11:384–400. doi: 10.1038/nrd3674. [DOI] [PubMed] [Google Scholar]
  • 88.Holliday R. Epigenetics: a historical overview. Epigenetics. 2006;1:76–80. doi: 10.4161/epi.1.2.2762. [DOI] [PubMed] [Google Scholar]
  • 89.Holoch D, Moazed D. RNA-mediated epigenetic regulation of gene expression. Nat Rev Genet. 2015;16:71–84. doi: 10.1038/nrg3863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Thiagalingam S, Cheng KH, Lee HJ, Mineva N, Thiagalingam A, Ponte JF. Histone deacetylases: unique players in shaping the epigenetic histone code. Ann N Y Acad Sci. 2003;983:84–100. doi: 10.1111/j.1749-6632.2003.tb05964.x. [DOI] [PubMed] [Google Scholar]
  • 91.Pang M, Zhuang S. Histone deacetylase: a potential therapeutic target for fibrotic disorders. The Journal of pharmacology and experimental therapeutics. 2010;335:266–272. doi: 10.1124/jpet.110.168385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hodges SJ, Yoo JJ, Mishra N, Atala A. The effect of epigenetic therapy on congenital neurogenic bladders--a pilot study. Urology. 2010;75:868–872. doi: 10.1016/j.urology.2009.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Qin L, Han YP. Epigenetic repression of matrix metalloproteinases in myofibroblastic hepatic stellate cells through histone deacetylases 4: implication in tissue fibrosis. Am J Pathol. 2010;177:1915–1928. doi: 10.2353/ajpath.2010.100011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ryu JK, Kim WJ, Choi MJ, Park JM, Song KM, Kwon MH, Das ND, Kwon KD, Batbold D, Yin GN, Suh JK. Inhibition of histone deacetylase 2 mitigates profibrotic TGF-beta1 responses in fibroblasts derived from Peyronie's plaque. Asian J Androl. 2013;15:640–645. doi: 10.1038/aja.2013.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kwon KD, Choi MJ, Park JM, Song KM, Kwon MH, Batbold D, Yin GN, Kim WJ, Ryu JK, Suh JK. Silencing histone deacetylase 2 using small hairpin RNA induces regression of fibrotic plaque in a rat model of Peyronie's disease. BJU international. 2014;114:926–936. doi: 10.1111/bju.12812. [DOI] [PubMed] [Google Scholar]
  • 96.Hefetz-Sela S, Stein I, Klieger Y, Porat R, Sade-Feldman M, Zreik F, Nagler A, Pappo O, Quagliata L, Dazert E, Eferl R, Terracciano L, Wagner EF, Ben-Neriah Y, Baniyash M, Pikarsky E. Acquisition of an immunosuppressive protumorigenic macrophage phenotype depending on c-Jun phosphorylation. Proc Natl Acad Sci U S A. 2014;111:17582–17587. doi: 10.1073/pnas.1409700111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Franceschi RT, Ge C, Xiao G, Roca H, Jiang D. Transcriptional regulation of osteoblasts. Cells Tissues Organs. 2009;189:144–152. doi: 10.1159/000151747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Liu Y, Zhang Y, Min J, Liu LL, Ma NQ, Feng YM, Liu D, Wang PZ, Huang DD, Zhuang Y, Zhang HL. Calcineurin promotes proliferation, migration, and invasion of small cell lung cancer. Tumour Biol. 2010;31:199–207. doi: 10.1007/s13277-010-0031-y. [DOI] [PubMed] [Google Scholar]
  • 99.Micutkova L, Diener T, Li C, Rogowska-Wrzesinska A, Mueck C, Huetter E, Weinberger B, Grubeck-Loebenstein B, Roepstorff P, Zeng R, Jansen-Duerr P. Insulinlike growth factor binding protein-6 delays replicative senescence of human fibroblasts. Mech Ageing Dev. 2011;132:468–479. doi: 10.1016/j.mad.2011.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES