Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Am J Physiol Endocrinol Metab 291: E952–E964, 2006.

First published June 6, 2006; doi:10.1152/ajpendo.00545.2005.

Mathematical model for the androgenic regulation of the prostate in intact and
castrated adult male rats
Laura K. Potter,1,2 Michael G. Zager,1,3 and Hugh A. Barton3
1
Curriculum in Toxicology, University of North Carolina, Chapel Hill; 2National Health and Environmental Effects
Research Laboratory, Experimental Toxicology Division, Office of Research and Development, United States
Environmental Protection Agency; and 3National Center for Computational Toxicology, Office of Research
and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina
Submitted 9 November 2005; accepted in final form 5 June 2006

Potter, Laura K., Michael G. Zager, and Hugh A. Barton. of prostate and other reproductive organs. In the pharmaceuti-
Mathematical model for the androgenic regulation of the prostate cal setting, therapeutic drugs such as finasteride, dutasteride,
in intact and castrated adult male rats. Am J Physiol Endocrinol bicalutamide, and flutamide are used to treat benign prostatic
Metab 291: E952–E964, 2006. First published June 6, 2006; hyperplasia and/or prostate cancer by inhibiting androgen-
doi:10.1152/ajpendo.00545.2005.—The testicular-hypothalamic- dependent growth processes.
pituitary axis regulates male reproductive system functions. Un-
The mechanisms of action of antiandrogens are generally of
derstanding these regulatory mechanisms is important for assessing
the reproductive effects of environmental and pharmaceutical an-
two forms. The first is the androgen antagonist, which binds to
drogenic and antiandrogenic compounds. A mathematical model the androgen receptor (AR) but does not stimulate DNA
for the dynamics of androgenic synthesis, transport, metabolism, transcription, such as the pharmaceutical compounds flutamide
and regulation of the adult rodent ventral prostate was developed and bicalutamide or the environmental compound vinclozolin.
on the basis of a model by Barton and Anderson (1997). The model The second is the 5␣-reductase inhibitor, which blocks the
describes the systemic and local kinetics of testosterone (T), metabolism of testosterone (T) to 5␣-dihydrotestosterone
5␣-dihydrotestosterone (DHT), and luteinizing hormone (LH), (DHT). The therapeutic drugs dutasteride and finasteride are
with metabolism of T to DHT by 5␣-reductase in liver and examples of 5␣-reductase inhibitors; no environmental exam-
prostate. Also included are feedback loops for the positive regu- ples are currently known. Both of these mechanisms effec-
lation of T synthesis by LH and negative regulation of LH by T and tively inhibit AR-mediated DNA transcription, leading to re-
DHT. The model simulates maintenance of the prostate as a duction of prostate size and cell numbers and decreased pros-
function of hormone concentrations and androgen receptor (AR)- tatic fluid production. Quantification of the male regulatory
mediated signal transduction. The regulatory processes involved in processes and their disruptions would aid in the dose-response
prostate size and function include cell proliferation, apoptosis,
assessment of environmental endocrine active compounds (4,
fluid production, and 5␣-reductase activity. Each process is con-
trolled through the occupancy of a representative gene by andro- 5). This requires biologically based quantitative methods de-
gen-AR dimers. The model simulates prostate dynamics for intact, scribing not only the pharmacokinetics and mode of action of
castrated, and intravenous T-injected rats. After calibration, the the exogenous compound of interest but also the pharmacoki-
model accurately captures the castration-induced regression of the netics of the endogenous hormones and how they regulate the
prostate compared with experimental data that show that the male reproductive organs.
prostate regresses to ⬃17 and 5% of its intact weight at 14 and 30 The objective in this research effort was to understand the
days postcastration, respectively. The model also accurately pre- normal adult male hormonal regulation of the prostate to set a
dicts serum T and AR levels following castration compared with foundation for quantitatively characterizing the effects of ex-
data. This model provides a framework for quantifying the kinetics posure to antiandrogens in male rats. To quantitatively under-
and effects of environmental and pharmaceutical endocrine active stand the normal adult male hormonal regulation of the pros-
compounds on the prostate. tate, mathematical descriptions of the kinetics of T, DHT, and
rodent ventral prostate; androgen receptor; testosterone; 5␣-dihy- luteinizing hormone (LH) are required, along with several
drotestosterone; testicular-hypothalamic-pituitary axis pharmacodynamic components, such as prostatic AR concen-
trations, maintenance of prostate size, and regulation of pros-
tatic fluid production.
THE HYPOTHALAMUS, PITUITARY, AND TESTES produce endocrine The regulation of T is crucial for prostate maintenance in
hormones responsible for regulation of the prostate and other that the size and function of the prostate is gene regulated via
male sexual functions (12, 46). Exogenous endocrine active DNA binding by androgen-bound AR (21–23, 37, 42, 53). T is
compounds can disrupt these processes. Some pesticides (e.g., produced mainly in the testicular Leydig cells in response to
vinclozolin and linuron) are known to have antiandrogenic LH released from the pituitary (12). It is metabolized to DHT
activity (19, 20). Toxic effects of antiandrogens in male ro- by 5␣-reductase in the liver and prostate (39, 50). The enzyme
dents range from developmental effects such as reproductive 5␣-reductase is upregulated by T and DHT via AR-mediated
malformations, retained nipples, and undescended testes to gene expression (39, 50). LH upregulates the production of T
pubertal effects such as delayed puberty and reduced weights and, hence, DHT, which in turn downregulates LH, creating a

Address for reprint requests and other correspondence: H. A. Barton, ORD The costs of publication of this article were defrayed in part by the payment
National Center for Computational Toxicology, US EPA, B205-01, Research of page charges. The article must therefore be hereby marked “advertisement”
Triangle Park, NC 27711 (e-mail: habarton@alum.mit.edu). in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

E952 http://www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION E953
negative feedback loop (12). In the blood, T and DHT bind to a single-compartment model is used for LH. Flow-limited compart-
serum albumin, which appears to play a role in regulating ments are utilized in cases where the uptake rate of a compound into
transport and tissue uptake dynamics (15, 33, 49). a tissue is limited by the blood flow rate into the tissue rather than the
In this article, a model describing the endogenous hormone diffusion rate of the compound across cell membranes. Conversely,
diffusion-limited compartments are used when the diffusion across
kinetics of the testicular-pituitary axis and the dynamics of the cell membranes is slow compared with the blood flow rate into the
androgenic regulation of the prostate is presented. The model tissue (32). The pharmacokinetics of T and DHT are modeled with
includes the pharmacokinetics of T, DHT, and LH, as well as compartments representing the systemic serum, liver, prostate, testes,
the dynamics of AR binding and signaling as they relate to the and rest of body (Fig. 1). LH is modeled with a single systemic
regulation of the prostate. Development of the model included compartment that captures androgen-regulated synthesis and the ef-
characterization of critical biological and physiological pro- fects of LH on T synthesis in the testes. All terms in the equations are
cesses inherent in the hormonal regulation of the male repro- defined in Tables 1, 2, 3, and 4.
ductive system, including androgen-LH feedback loops and Systemic serum compartment. The compartment for systemic se-
AR-mediated regulation of the prostate. rum includes exchange of T and DHT with the tissue compartments,
The model structure for the hormonal transport, or pharma- the binding dynamics of T and DHT to albumin, and a basal input rate
of T, due to limited synthesis of T outside the testes (29). A
cokinetic component, is based on standard physiologically simplifying assumption is made that all concentrations are rapidly
based pharmacokinetic (PBPK) modeling (32), whereas the equilibrated between red blood cells and serum.
dynamics of the AR-based regulation of the prostate are based The rate equation for the kinetics of T in the systemic serum is
on mass action kinetics. These dynamics include binding of T given as
and DHT to the AR, dimerization of androgen-AR complexes,
dAT bl
and gene activation by these complexes via DNA binding. The ⫽ Qb共CTbf ⫺ CTblf兲 ⫹ Qp共CTpf ⫺ CTbl兲 ⫹ Ql共CTlf ⫺ CTbl兲
complexes regulate prostatic cellular mass, fluid production, dt
(1)
blood flow, AR concentrations, and 5␣-reductase activity. ⫹ Qt共1 ⫺ ts兲共CTtv ⫺ CTblf兲 ⫹ k2T
Unknown physiological, kinetic, and binding parameters The first four terms represent T exchange with the rest of body,
were estimated using available experimental data from the prostate, liver, and testes compartments, respectively, followed by the
literature. The resulting calibrated model is capable of quanti- basal synthesis of T. Free concentrations CTblf are affected by albu-
tatively describing intact and castrated rats at steady state, as min binding in the blood as in Eq. 3 below. It is assumed that only free
well as the dynamics that occur following castration. This T is available to enter the testes and rest-of-body compartments,
model may be used for testing and generating hypotheses, data whereas both free and albumin-bound T are available for uptake into
interpretation, experimental design, and identifying key data the metabolically active prostate and liver (33).
gaps in the study of the hormonal regulation of the prostate and The equation for the kinetics of DHT in systemic serum has a
provides a framework for elaborations that address the effects similar form, except that basal DHT synthesis is not included, because
of exposure to exogenous, pharmaceutical, or environmental it is assumed that all significant amounts of DHT are produced in the
prostate and liver via metabolism (1). In addition, the rest-of-body
androgens and antiandrogens (5). compartment for DHT incorporates the testes, which is modeled as a
separate compartment for T to capture the dynamics of T synthesis.
METHODS The equation for DHT transport in the systemic serum is given by
Model Structure: Pharmacokinetic Component dAD bl
⫽ Qbt共CDbf ⫺ CDblf兲 ⫹ Qp共CDpf ⫺ CDblf兲 ⫹ Ql共CDlf ⫺ CDbl兲
The transport and dispositional kinetics of T and DHT are de- dt
scribed using standard PBPK compartmental modeling (32), whereas (2)

Fig. 1. Model schema for kinetics of testostwerone (T) and


5␣-dihydrotestosterone (DHT). Arrows depict blood flow to
and from tissue compartments, metabolism and clearance. Syn-
thesis of T occurs in blood and testicular Leydig cells (LC),
with androgen binding to albumin in the blood and to androgen
receptor (AR) in the prostate. The components of the testes
compartment for testosterone include the spermatic cord venous
blood (SV), testicular venous blood (TV), interstitial fluid (IF),
and seminiferous tubules (ST). Blood flow parameters are given
in Table 2.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
E954 MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION

Table 1. Model variables


Variable Symbol Initial Value Reference

Concentration of LH CLH 0.104 (6)


Concentration of T in testicular interstitial fluid CTif 307.29 (6)
Concentration of T in testicular seminiferous tubules CTst 307.29 (6)
Concentration of T in testicular venous blood CTtv 153.92 Steady state
Concentration of T in prostate CTp 11.38 (31)
Concentration of DHT in prostate CDp 52.46 (42)
Free concentration of AR in prostate CRf 0.95 Steady state
Concentration of T-bound AR in prostate CT:R 3.82 Steady state
Concentration of DHT-bound AR in prostate CD:R 15.76 Steady state
Concentration of dimerized complex T:AR-T:AR in prostate CTT 0.65 Steady state
Concentration of dimerized complex DHT:AR-T:AR in prostate CDT 2.69 Steady state
Concentration of dimerized complex DHT:AR-DHT:AR in prostate CDD 11.11 Steady state
Concentration of dimer T:AR-T:AR bound to cd DNA in prostate (cell death) CTTcd 3.35 ⫻ 10⫺4 Steady state
Concentration of dimer T:AR-T:AR bound to sec DNA in prostate (fluid secretion) CTTsec 3.16 ⫻ 10⫺4 Steady state
Concentration of dimer T:AR-T:AR bound to cp DNA in prostate (cell proliferation) CTTcp 2.48 ⫻ 10⫺4 Steady state
Concentration of dimer T:AR-T:AR bound to 5a DNA in prostate (5␣-reductase) CTT5a 3.41 ⫻ 10⫺4 Steady state
Concentration of dimer DHT:AR-DHT:AR bound to cd DNA in prostate CDDcd 0.068 Steady state
Concentration of dimer DHT:AR-DHT:AR bound to sec DNA in prostate CDDsec 0.065 Steady state
Concentration of dimer DHT:AR-DHT:AR bound to cp DNA in prostate CDDcp 0.051 Steady state
Concentration of dimer DHT:AR-DHT:AR bound to 5a DNA in prostate CDD5a 0.070 Steady state
Concentration of dimer T:AR-DHT:AR bound to cd DNA in prostate CDTcd 0.0021 Steady state
Concentration of dimer T:AR-DHT:AR bound to sec DNA in prostate CDTsec 0.0020 Steady state
Concentration of dimer T:AR-DHT:AR bound to cp DNA in prostate CDTcp 0.0015 Steady state
Concentration of dimer T:AR-DHT:AR bound to 5a DNA in prostate CDT5a 0.0021 Steady state
Free concentration of cd DNA binding site CDNAcdf 0.0044 Steady state
Free concentration of sec DNA binding site CDNAsecf 0.0083 Steady state
Free concentration of cp DNA binding site CDNAcpf 0.023 Steady state
Free concentration of 5a DNA binding site CDNA5af 0.0031 Steady state
Concentration of T in liver CTl 9.92 Steady state
Concentration of DHT in liver CDl 0.92 Steady state
Concentration of T in systemic serum CTbl 7.60 (6)
Concentration of DHT in systemic serum CDbl 0.59 (31)
Concentration of T-bound albumin in systemic serum CT:A 6.97 Steady state
Concentration of DHT-bound albumin in systemic serum CD:A 0.56 Steady state
Free concentration of albumin in systemic serum CAf 499992 Steady state
Occupancy of cd DNA binding site DNAocd 0.94 Steady state
Occupancy of sec DNA binding site DNAosec 0.90 Steady state
Occupancy of cp DNA binding site DNAocp 0.70 Steady state
Occupancy of 5a DNA binding site DNAo5a 0.96 Steady state
Volume of androgen-sensitive prostatic cell mass VPCl 1.00 ⫻ 10⫺4 (42)
Volume of androgen-sensitive prostatic ductal lumen mass VPLl 1.79 ⫻ 10⫺4 (42)
Total volume of prostate Vp 3.30 ⫻ 10⫺4 (52)
Concentrations are given in nM, volumes in liters, and occupancy is unitless. Equations in the text denote amounts with A (e.g., ATif is amount of T in testicular
interstitial fluid), and free tissue concentrations of T and DHT are denoted with f appended to the subscript (e.g., CTpf is free concentration of T in prostate).
Initial conditions for intact steady state are given in the 3rd column, with references in the 4th column. Initial values with the steady-state reference were
determined by matching intact and castrate steady states with experimental data when available (see Table 5) or by running the model to steady state after
calibration.

We assume standard binding kinetics for T and DHT binding to Michaelis-Menten term, and linear terms are included for the general
albumin: metabolic clearance of T and DHT in the liver (49). The rate equations
for T and DHT in the liver are given by
dCT:A
⫽ kTAonCTblfCAf ⫺ kTAoffCT:A (3)
dt dAT l VmaxlCTlf ATl
⫽ Ql共CTbl ⫺ CTlf兲 ⫺ ⫺ klT (8)
dCD:A dt km5␣ ⫹ CTlf PTl
⫽ kDAonCDblfCAf ⫺ kDAoffCD:A (4)
dt and
with free T, DHT, and albumin concentrations determined using mass dAD l VmaxlCTlf ADl
balance: ⫽ Ql共CDbl ⫺ CDlf兲 ⫹ ⫺ klD (9)
dt km5␣ ⫹ CTlf PDl
CT blf ⫽ CTbl ⫺ CT:A (5)
Free concentrations of T and DHT in the liver are given by the
CD blf ⫽ CDbl ⫺ CD:A (6) algebraic equations
CA f ⫽ CA ⫺ CT:A ⫺ CD:A (7)
CTl CDl
CT lf ⫽ , CDlf ⫽ (10)
Liver compartment. The kinetics of T and DHT in the liver are PTl PDl
described by using a flow-limited compartmental model. Metabolism
of T to DHT via 5␣-reductase in the liver is modeled with a so that free concentrations are proportional to total concentrations.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION E955
Table 2. Model physiological parameters
Parameter Symbol Value Reference

Rat body mass bm 0.3 kg (6)


Rat body volume bv 0.3 liters Assuming density ⫽ 1
Volume of blood Vbl 0.021 liters (6)
Volume of liver Vl 0.012 liters (7)
Volume of rest-of-body (ROB) excluding testes Vb 0.24 liters (6)
Volume of ROB including testes Vbt 0.25 liters (6)
Volume of testes Vt 0.0033 liters (6)
Volume of interstitial tissue Vit 3.63 ⫻ 10⫺4 liters (6)
Volume of seminiferous tubules Vst 0.0029 liters (6)
Volume of interstitial fluid Vif 2.25 ⫻ 10⫺4 liters (6)
Basal prostatic ductal lumen volume VPL2 3.08 ⫻ 10⫺6 liters (42)
Basal prostate cell volume VPC2 5.08 ⫻ 10⫺5 liters (42)
Cardiac output Qc 6.08 l/h (6)
Steady state blood flow to prostate Qp0 0.024 l/h (16)
Proportional constant for Qp kQp 73.84 h⫺1 Steady state
Blood flow rate to testes (intact only) Qt 0.061 l/h (6)
Blood flow rate to liver Ql 1.06 l/h (7)
Blood flow rate to ROB excluding testes Qb Qc-Qp-Qt-Ql Flux balance
Blood flow rate to ROB including testes Qbt Qc-Qpt-Ql Flux balance
Fraction of testicular blood flow shunted to SV ts 0.56 (6)
Total DNA concentration per site (k ⫽ cp, cd, sec, 5a) CDNAk 0.075 nM Fitted
Total concentration of serum albumin CA 5.00 ⫻ 105 nM (10)
Parameters with steady state reference were determined by running the calibrated model to steady state.

Testes compartment. The testes compartment has a unique structure the blood flow directly into the spermatic cord (bypassing the testes),
based on the specific biology of the testes that is important for T since the blood flow rate to the testes compartment is split into two
the testicular Leydig cells are the major source for T synthesis (12). components (Fig. 1). One component of the blood flow feeds into the
Here, we give a brief outline of the model; for a comprehensive IF subcompartment, and the other component flows into the spermatic
development of the model structure and equations, the reader is cord and feeds directly into the systemic serum. The bias of the split
referred to Barton and Anderson (6). As previously mentioned, the is controlled by the model parameter ts.
kinetics of DHT in the testes are incorporated in the rest-of-body Circulating T reenters the IF via the blood and then diffuses
compartment, so that the testes compartment is utilized only for T. between the IF and ST as with a standard diffusion-limited compart-
The testes compartment is divided into two subcompartments, ment (32). The ST compartment is present to facilitate future elabo-
representing the seminiferous tubules (ST) and the interstitial fluid rations to address spermatogenesis. The diffusion of T between the IF
(IF). To account for the testicular shunt (30) that directs a portion of and ST is controlled in the model by the parameter ␮. To account for

Table 3. Model hormone pharmacokinetic parameters


Parameter Symbol Value Reference

IF/blood partition coefficient for T PTif 1.96 Fitted


ST/IF partition coefficient for T PTst 1.00 (6)
Liver/blood partition coeff. for T PTl 2.75 Fitted
Liver/blood partition coeff. for DHT PDl 2.00 Fitted
ROB/blood partition coeff. for T PTb 1.00 (6)
ROB/blood partition coeff. for DHT PDb 0.62 Fitted
Testicular permeability coefficient ␮ 1000 l/h (6)
Coeff. for nonspecific T binding in prostate NTp 0.79 Fitted
Coeff. for nonspecific DHT binding in prostate NDp 1.00 Fitted
Relative potency of T for LH synthesis inhibition ␣T 0.25 (55)
Relative potency of DHT for LH synthesis inhibition ␣D 0.75 (55)
T synthesis rate in testes (intact only) k1t 40.32 l/h Fitted
Basal T synthesis rate k2t 0.17 nmol/h Fitted
Basal AR synthesis rate k1R 68.15 nmol/h Fitted
T elimination rate in liver k1T 87.93 h⫺1 Fitted
DHT elimination rate in liver klD 77.20 h⫺1 Fitted
AR elimination rate keR 71.90 l/h Fitted
T inhibition constant 1 for LH kLH1 0.13 l/h Fitted
T inhibition constant 2 for LH kLH2 0.026 nmol/h Fitted
Minimum LH synthesis rate kLH3 1.68 ⫻ 10⫺4 nmol/h Fitted
LH degradation rate constant kLH4 0.80 h⫺1 Fitted
Regulation constant for Vmax in prostate k5a 3.02 nmol/h Steady state
Vmax for T metabolism to DHT in liver Vmax1 3.65 nmol/h Fitted
Steady state Vmax for T metabolism to DHT in prostate Vmax0 2.89 nmol/h Fitted
km for T metabolism to DHT in prostate km5a 40.00 nM (35)
Initial values with the steady state reference were determined by running the calibrated model to steady state.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
E956 MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION

Table 4. Model biochemical and kinetic parameters for albumin, AR, DNA, and prostate
Parameter Symbol Value Reference

Association rate for T-AR kTRon 0.14 nM⫺1 h⫺1 (53)


Dissociation rate for T-AR kTRoff 0.069 h⫺1 Computed from (53)
Association rate for DHT-AR kDRon 0.053 nM⫺1 h⫺1 (53)
Dissociation rate for DHT-AR kDRoff 0.018 h⫺1 Computed from (53)
Equilibrium dissociation constant for T-albumin KTAd 4.50 ⫻ 104 nM (49)
Association rate for T-albumin kTAon 0.18 nM⫺1 h⫺1 Fitted
Dissociation rate for T-albumin kTAoff 8131 h⫺1 kTAoff ⫽ kTAon KTAd
Equilibrium dissociation constant for DHT-albumin KDAd 2.89 ⫻ 104 nM (49)
Association rate for DHT-albumin kDAon 0.15 nM⫺1 h⫺1 Fitted
Dissociation rate for DHT-albumin kDAoff 5407 h⫺1 kDAoff ⫽ kDAon KDAd
Association rates for androgen dimerization (I and J represent the 2
androgens in the dimer) kIJon 0.14 nM⫺1 h⫺1 Assumption
Dissociation rates for androgen dimerization kIJoff 3.13 h⫺1 Fitted
Association rates for DNA dimer formation (I and J represent the 2 dimers;
k denotes the representative genes (cd, cp, 5a, sec) kDNAonIJk 0.14 nM⫺1 h⫺1 Assumption
Dissociation rates for DNA dimer formation kDNAoffIJk See text Fitted
Steady state DNA occupancy for 5␣-reductase activity DNAo5a0 0.96 Steady state
Steady state DNA occupancy for cell proliferation DNAocp0 0.70 Steady state
Steady state DNA occupancy for cell death DNAocd0 0.94 Steady state
Steady state DNA occupancy for fluid production DNAosec0 0.89 Steady state
Fluid outflow rate constant kflo 0.043 h⫺1 Fitted
Fluid production rate constant ksec 8.44 ⫻ 10⫺6 kg/h Steady state
Prostate cell proliferation rate constant kcpl 1.17 ⫻ 10⫺7 kg/h Fitted
Prostate cell death rate constant kcdl 0.014 h⫺1 Steady state
Parameters with steady state reference were determined by running the calibrated model to steady state.

the synthesis of T in the Leydig cells, there is a source of T production dAD p VmaxCTpf
in the IF (12). Recalling that LH upregulates T synthesis in the Leydig ⫽ Qp共CDblf ⫺ CDpf兲 ⫹ (15)
cells (6), we assume a positive linear feedback mechanism in the dt km5␣ ⫹ CTpf
model with rate constant k1T. Once the testicular venous blood leaves Free prostatic T and DHT are available to bind to AR. The androgen-
the testes compartment, it immediately reenters the systemic serum bound AR complexes T:AR and DHT:AR then form homo- and het-
via the spermatic cord. The rate equation for T in the IF subcompart- erodimers. These dimers bind to DNA and stimulate the regulation of various
ment is given by processes. The equations for T and DHT binding to AR are given by
dAT if
dt

⫽ Qt共1 ⫺ ts兲 CTblf ⫺
CTif
PTif
⫹␮冊 冉
CTst
PTst

⫺ CTif ⫹ klTCLH (11)
dCT:R
dt
⫽ kTRonCTpfCRf ⫺ kTRoffCT:R (16)

With only diffusion occurring in the ST compartment, the correspond- and


ing rate equation is defined as dCD:R
⫽ kDRonCDpfCRf ⫺ kDRoffCD:R
冉 冊
(17)
dAT st CTst dt
⫽ ␮ CTif ⫺ (12)
dt PTst

The algebraic equation for the testicular venous blood describes the
concentration of T as proportional to the concentration in the inter-
stitial fluid due to nonspecific binding and is given by
CTif
CT tv ⫽ (13)
PTif
Prostate compartment. The prostate is a complex tissue with
multiple lobes and multiple cell types, which vary in their androgen
responsiveness (41). The model currently is parameterized to describe
the ventral lobe of the prostate, which represents 60 –70% of the rat
prostate (11). The prostate compartment includes transport to and
from the blood, the metabolism of T to DHT by 5␣-reductase, and the
dynamics of AR binding, dimerization, and DNA binding (Fig. 2).
The rate equations for the kinetics of T and DHT in the prostate are
given (respectively) by
dAT p VmaxCTpf Fig. 2. Schema of androgenic regulation in the prostate. T is converted to
⫽ Qp共CTbl ⫺ CTpf兲 ⫺ (14) DHT, either of which can bind to the AR and form homogeneous or hetero-
dt km5␣ ⫹ CTpf geneous dimers (T binding not shown here). AR-mediated DNA transcription
leads to upregulation of 5␣-reductase, cell proliferation, and fluid production
and as well as downregulation of apoptosis.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION E957
The homo- and heterodimers (abbreviated as DT, DD, and TT) have Rest-of-body compartment. The rest-of-body compartment repre-
similar kinetics: sents all tissues and organs not included in the remaining compart-
ments and includes exchange with the systemic serum. Note that the
dCDT testes are included in the rest-of-body compartment for DHT but not
⫽ kDTon共CD:R兲共CT:R兲 ⫺ kDToffCDT (18)
dt for T since this tissue is modeled separately for T synthesis dynamics.
This results in two different blood flow rates, Qb and Qbt for the
dCDD
⫽ kDDon共CD:R兲2 ⫺ kDDoffCDD (19) rest-of-body compartments that exclude and include the testes. The
dt transport equations for T and DHT in the rest of body are given by
dCTT
dt
⫽ kTTon共CT:R兲2 ⫺ kTToffCTT (20)
dAT b
dt

⫽ Qb CTblf ⫺
CTb
PTb
冊 (27)

The dimers above are able to bind to DNA sites to form DNA-dimer and

冉 冊
complexes. In the model, there are four distinct DNA binding sites
that are involved in regulating 5␣-reductase synthesis, cell prolifera- dAD b CDb
⫽ Qbt CDblf ⫺ (28)
tion, apoptosis, and prostatic fluid production. Although numerous dt PDb
genes are likely responsible for the regulation of the last three
processes, in the model the simplifying assumption was made to LH pharmacokinetics. The kinetics of LH are described in the
represent each process by a single gene. model with a single equation that incorporates the synthesis and
The four representative genes in the model are available to bind to elimination of LH in the serum. The synthesis of LH is negatively
all three dimer types, yielding 12 equations for the concentrations of regulated by T and DHT concentrations, with a small basal amount of
the DNA-dimer complexes. One example is given here. The following synthesis assumed. Similar to the upregulation of T by LH, we assume
equation is for the concentration of the DHT:AR-DHT:AR dimer a piecewise linear feedback mechanism to describe downregulation of
bound to the 5␣-reductase synthesis-regulating gene: LH by T. Elimination of LH is proportional to the amount of LH in the
system. The transport equation is given by
dCDD 5a
⫽ kDNAonDD5aCDD 䡠 CDNA5af ⫺ kDNAoffDD5aCDD5a (21) dALH
dt ⫽ max [⫺kLH1共␣TCTblf ⫹ ␣DCDblf兲 ⫹ kLH2,kLH3] ⫺ kLH4ALH
dt
where parameters and variables for the 12 DNA-dimer complexes are (29)
given in Tables 1, 2, and 4. The dissociation rates for the DNA-dimer
complexes are determined to reflect relative potencies between T and where ␣T and ␣D represent the relative potency for inhibiting LH
DHT as well as between the four representative DNA binding sites. production, kLH1 and kLH2 describe the decline in LH with increasing
Specifically, we assume that the relative potencies for the dissociation androgen, kLH3 denotes the minimum LH synthesis rate, and kLH4
of DNA from the dimers T:AR-T:AR, D:AR-D:AR, and D:AR-T:AR represents LH degradation. See Fig. 3 for a representative graph of the
are 6, 0.5, and 4, respectively. These potencies account for the fact LH synthesis rate function.
that DHT is significantly more potent in mediating AR-based effects
than T is (55). Moreover, the four representative DNA binding sites Model Structure: Pharmacodynamic Component
have relative potencies for dissociation with DNA-dimer complexes The model describes the dynamics of AR concentrations, prostatic
that reflect the differential responses of the four regulatory events blood flow, 5␣-reductase activity, prostatic ductal lumen mass, and
(5␣-reductase activity, fluid production, cell proliferation, and apop- prostate cellular mass. Prostatic blood flow is regulated by the size of
tosis) to AR-mediated signaling. These potencies are 0.14, 0.4, 1.4, the prostatic cellular mass, whereas 5␣-reductase activity, cellular
and 0.2, respectively, and were estimated as part of the parameter mass, and ductal lumen mass are regulated via occupancy of DNA
estimation process described below. The model is formulated so that binding sites.
the occupancy of each gene can hold any value between 0 and 1,
representing the fraction of total possible occupancy.
Free concentrations of prostatic T, DHT, AR, and DNA sites are
given by

CT pf ⫽
CTp ⫺ CT:R ⫺ 2CTT ⫺ CDT ⫺ 兺 共T兲 (22)
1 ⫹ NTp

CD pf ⫽
CDp ⫺ CD:R ⫺ 2CDD ⫺ CDT ⫺ 兺 共D兲 (23)
1 ⫹ NDp

CR f ⫽ CR ⫺ CT:R ⫺ CD:R ⫺ 2 兺 共dimer兲 ⫺ 2兺 共DNA兲 (24)

and
CDNA if ⫽ CDNAj ⫺ 共CTTj ⫹ CDDj ⫹ CDTj兲, j ⫽ cd,sec,cp,5a
(25)
where NTp and NDp are coefficients for nonspecific binding of T and
DHT, respectively, in the prostate, and ¥(T) and ¥(D) are all the
Fig. 3. LH synthesis rate function. LH synthesis is negatively regulated by
DNA-bound dimers containing T and D, respectively. The equations androgens and is modeled as a piecewise linear function with a minimal basal
for DNA occupancy are given by synthesis rate KLH LH LH
3 . Parameters K1 and K2 represent the slope and y-intercept
(respectively) of the leftmost linear piece. Weighted free androgen concentra-
CTTj ⫹ CDDj ⫹ CDTj
DNAo j ⫽ , j ⫽ cd,sec,cp,5a (26) tion reflects the stronger relative potency of DHT vs. T for LH synthesis
CDNAj inhibition. Parameters are detailed in Table 4.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
E958 MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION

AR concentrations. Although the AR has been shown to be up- fluid production gene in the model. The governing equation is given
regulated by androgens (48), here we make the simplifying assump- by
tion that prostatic AR is produced at a constant rate and free AR is
eliminated at a linearly proportional rate. We further assume that dVPL 1 VPC1共t兲
⫽ ksecDNAosec ⫺ kfloVPL1 (34)
androgen-bound AR is not available for elimination. The equations for dt VPC1b
total AR amount and concentration, respectively, are as follows:
The growth rate of the ductal lumen is proportional to the occupancy
dAR AR DNAosec of the representative fluid production gene and the size of
⫽ k1R ⫺ keRCRf, CR ⫽ (30)
dt Vp the androgen-sensitive prostate cellular mass relative to the intact,
steady-state cellular mass VPC1b. Specifically, when cellular mass
Recall that the concentration of free prostatic AR is given in Eq. 24. decreases following castration or antiandrogen exposure, the rate of
Prostatic blood flow. Although it has been shown that prostatic fluid production concurrently decreases, since there are fewer cells
blood flow decreases almost immediately after castration (45), here present to secrete fluid. We also assume that there is linear degrada-
we make the simplifying assumption that the blood flow to the tion of ductal lumen as fluid exits the prostate.
prostate is proportional to the volume of the prostatic cell mass Vpc: The rate constants for cell proliferation and apoptosis in Eq. 33
Q p ⫽ kQpVpc (31) were chosen so that both terms of the equation are equal when the
model reaches intact steady-state values, thus yielding equilibrium
5␣-Reductase activity. The activity of 5␣-reductase and, hence, the and no change in prostate cellular mass. The rate constants for fluid
metabolism of T are regulated by AR binding and signaling (18, 43). secretion and fluid flow in Eq. 34 were chosen similarly so that the
Here, we assume that Vmax is directly proportional to the occupancy, entire ventral prostate is at equilibrium when steady-state values are
DNAo5a, of the representative 5␣-reductase gene: reached.
Note that in Eqs. 33 and 34, the ratio VPC1(t)/VPC1b can become
V max ⫽ k5aDNAo5a共t兲 (32)
unbounded under conditions of supernormal hormone levels and
Androgen-sensitive prostatic cellular mass. On the basis of exper- prostate growth. However, in the scope of these modeling efforts,
imental data (11, 42), we assume that the prostate contains a basal supernormal hormone levels will not be simulated, and therefore
cellular mass, VPC2, that is not affected by androgens and an andro- supernormal prostate volumes are not expected. If one were to use the
gen-sensitive cellular mass that is regulated via the occupancy of the model under conditions of supernormal hormone levels, it would be
representative cell proliferation and apoptosis genes (Fig. 4). The necessary to address this limitation, likely using experimental data
equation for the volume of the androgen-sensitive cellular mass is as from androgen implant experiments in intact and castrated rats to
follows: define an appropriate function.
Algebraic equation for prostate volume. Adding the basal and
dVPC 1 VPC1共t兲 androgen-sensitive volumes of the prostate cellular mass and ductal
⫽ kcp1DNAocp ⫺ kcd1共1 ⫺ DNAocd兲VPC1共t兲 (33)
dt VPC1b lumen mass in the model, we are able to compute the total volume of
the prostate as a function of time:
This is based on the assumption that the proliferation rate is propor-
tional to the occupancy DNAocp of the representative cell prolifera- V p共t兲 ⫽ VPC1共t兲 ⫹ VPC2 ⫹ VPL1共t兲 ⫹ VPL2 (35)
tion gene and that the rate of cell formation increases with prostate
cellular mass. Moreover, the death rate is negatively related to the Model Implementation and Calibration
occupancy DNAocd of the representative apoptosis gene, since in this
case DNA occupancy results in protection from apoptosis (36). The model was implemented computationally in Matlab (The
Androgen-sensitive prostatic ductal lumen mass. As with the pros- Mathworks, Natick, MA). Parameters for the model were chosen on
tate cellular mass, we assume that there is a basal amount VPL2 of the the basis of values from the literature where possible. Other parameter
ductal lumen that remains unaffected by androgen concentrations and values were estimated by comparison with experimental data, and a
an androgen-sensitive amount that is regulated by the representative selected set was reestimated using standard least squares parameter
estimation techniques (51).
One parameter estimation problem was formulated to estimate
pharmacokinetic parameters such as tissue/blood partition coefficients
and the feedback parameters for LH and T synthesis. These parame-
ters were estimated using a submodel that contains only the pharma-
cokinetics of T, DHT, and LH without any of the pharmacodynamic
phenomena. The estimation problem involved comparing submodel
simulations to intact and castrate steady-state values and data as in (6).
A second parameter estimation problem was formulated with the
full model to estimate pharmacodynamic parameters, such as binding
constants, prostatic fluid production rate, 5␣-reductase Vmax, and
prostatic cell proliferation and death rates. These parameters were
estimated in comparison with intact and castrate steady-state behav-
iors and the dynamics of prostate regression following castration.
The third and final estimation problem was formulated with the full
model and included all parameters estimated in the first two estima-
tion problems, where the initial value for each unknown parameter
was the optimal value obtained from the respective previous estima-
tion problem. This allowed for a more accurate fit to the overall
model, since a submodel was used in the first problem to estimate
Fig. 4. Prostate cell mass and duct lumen subcompartments. Basal cell mass reasonable starting points for the pharmacokinetic parameters. Model
and duct lumen subcompartments have constant volumes, whereas androgen- fits using the final parameter values are presented in RESULTS. A
sensitive cell mass subcompartments are regulated by androgens and vary with complete list of model parameters with values and references is given
time. Model variables and parameters are detailed in Tables 1 and 2. in Tables 2– 4.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION E959
Table 5. Comparison of model-simulated intact and castrate
steady-state values with experimental data
Intact Steady State Castrate Steady State

Hormone Model Experimental data Model Experimental data

Serum T, nM 7.6 7.6 (6) 0.31 See RESULTS


Free serum T, % 8.3 22.0 (49) 8.3 NA
Prostatic T, nM 11.4 12.1 (31) 1.13 NA
Free prostatic T, % 17.4 6.7 (55) 26.2 NA
T in IF, nM 307.3 307.0 (6)
Serum DHT, nM 0.59 0.6 (31) 0.01 0.07 (17)
Free serum DHT, % 5.5 18.0 (49) 5.46 NA
Prostatic DHT, nM 52.5 53 (42) 0.09 NA
Free prostatic DHT, % 10.8 NA 19.2 NA
Serum LH, nM 0.104 0.104 (6) 1.49 NA

RESULTS
Fig. 6. Effects of castration on serum T. Model simulations (solid line) are
The model was used to predict the dynamics of androgen compared with experimental data as a fraction of intact values.
levels and prostate regulation under various conditions, includ-
ing the intact steady state, exogenous dosing of T via intrave-
nous injections, and castration. The simulations reported in this tions, followed by decaying oscillatory effects due to the
section vary in time duration from 8 h to 30 days. Although rats stabilizing ability of the feedback system. After ⬃4 h from the
grow significantly in the time span of 30 days and the prostate time of injection, the system is essentially stable.
grows at a similar rate to the whole body (8, 37), we assume a
constant body mass in the model and report all simulations and Simulating Serum T Levels After Castration
experimental data as fractions of intact values. Systemic T concentrations drop rapidly following castration
Predicting Steady-State Concentrations (9, 26). Data show that, within the first 30 min, serum T levels
drop to ⬃15% of intact values. After 6 h, serum T levels are
Table 5 reports steady-state concentrations of T, LH, and reported to be less than 5% of intact values. Figure 6 depicts
DHT in serum and various tissues for intact and castrate these data and the corresponding model prediction. Serum T in
conditions. These concentrations were obtained by simulating castrated rats is often close to assay detection limits and is quite
an unperturbed intact or castrate state until equilibrium was variable, as evidenced by values ranging from 0.4 to 5% of
obtained for all model species. The results given in Table 5 intact values in Kyprianou and Isaacs (26). Therefore, an
suggest that the model is capable of predicting both intact and extratesticular synthesis rate of T included in the systemic
castrate steady-state concentrations of T, DHT, and LH com- serum compartmental model was calibrated to give 5% of
pared with the data. intact levels, which yielded a good overall fit with all of the
experimental data used to calibrate the model.
Simulating an Intravenous Dose of T
Simulating Regression of the Prostate Postcastration
Results of a simulated intravenous injection of 0.02 nmol T
are shown in Fig. 5, which depicts time courses for T and LH Suzuki et al. (48) report substantial decreases in prostatic
concentrations in blood and T concentrations in the testes. The AR levels in response to castration. Figure 7 depicts the model
plots illustrate the ability of the model to simulate T and LH prediction vs. the experimental data. As can be seen, the
regulation in serum and the testes after a perturbation of the predicted AR levels at each time point are within the given
intact steady-state system. Note the immediate spike in serum error bars for the data.
T concentrations from the steady-state concentration of 7.6 Several studies have demonstrated a sigmoidal decrease in
nM, and the resulting acute decrease in serum LH concentra- prostate weight in time after castration (27, 28, 40, 42). Figure

Fig. 5. Simulated T and LH concentrations following iv


injection with 0.02 nmol T. After initial perturbation of
the steady-state system (serum T 7.6 nM, serum LH
0.10 nM), feedback between T and LH forces the
system back to near-equilibrium within 4 h. The imme-
diate spike in serum T due to the injection causes an
immediate drop in LH levels, resulting in an acute
decrease in T production in the testes, demonstrated by
the drop in concentration depicted. Resulting waves in
plots are due to feedback mechanisms forcing the sys-
tem to equilibrium.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
E960 MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION

Fig. 9. Effects of castration on prostatic ductal lumen mass. Model simulations


Fig. 7. Effects of castration on prostatic AR levels. Model simulations (solid (solid line) are compared with experimental data.
line) are compared with experimental data as a fraction of intact values.

8 includes model predictions of postcastration prostate weight slower pace, reaching its minimum ⬃2 wk after castration.
vs. experimental data. The model predictions match well with These dynamics, which are shown together with corresponding
a majority of the data across the different experiments. More- model simulations in Fig. 10, are consistent with a gene-
over, the simulations suggest that the prostate has lost all regulatory delay in the prostatic response to androgen removal.
androgen-sensitive cellular mass and ductal lumen mass by Model Validation
about the 11th day after castration. The remaining androgen-
insensitive prostate mass is ⬃16.4% of the total intact mass. The predictive accuracy of the model was tested by com-
In addition to predicting the time course of the entire paring simulations with experimental data that were not used in
prostate volume after castration, model simulations capture the the calibration process. These include prostatic blood flow rate
regression of the ductal lumen component. Figure 9 depicts measurements after castration as well as additional data for
model predictions of the ductal lumen mass after castration postcastration regression of the prostate. Figure 11 depicts
compared with experimental data (42), showing a good match model simulations compared with the additional prostate
between the data and the model. weight and blood flow data. The model accurately predicts the
As illustrated by the experimental data presented in Figs. prostate weight data, although it overpredicts the weight at 72 h
6 –9, there is a clear sequence of regulatory events that occurs postcastration. The data from these two studies at this partic-
after castration, leading to decreased prostate weight. Within ular time point happen to be significantly lower than data at the
1 h, there is a sharp decrease in serum androgen concentrations same point from the studies used to calibrate the model (see
followed by a drop in prostatic AR levels over a couple of Fig. 8), which explains why the model is overpredicting the
days. As AR concentrations decrease, the prostate responds new data.
more slowly with decreased fluid production, so that the ductal Moreover, the model predictions for prostatic blood flow
lumen mass shrinks to less than 10% of the intact mass within rate after castration are reasonably accurate compared with the
a week. Finally, the prostatic cellular mass regresses at a data (Fig. 11). Note that no prostatic blood flow data were used
in the model calibration process, so the accurate prediction of
prostatic blood flow helps to further validate the model as a
plausible representation of the biology. Because the current
state of the model approximates the relationship between blood
flow and tissue volume without describing the complex causal
relationship (24, 36a), these results may further suggest that the
simplified description provides a reasonable approximation of
prostatic blood flow behavior.
DISCUSSION

The objective of this research effort was to understand the


normal adult male hormonal regulation of the prostate as a
foundation for quantitatively characterizing the effects of en-
vironmental or pharmaceutical antiandrogen exposure in male
rats. Because it is impossible to capture all physiological and
biological details in a mathematical model, a typical approach
is to describe the critical determinants of biological behavior
while reasonably simplifying many other details. The result is
Fig. 8. Effects of castration on prostate weight. Model simulations (solid line) a model that is a mix of mechanistic and empirical elements.
are compared with experimental data as a fraction of intact values. The goal is then to obtain a parameter set that produces
AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION E961

Fig. 10. Timing of postcastration events. Model simu-


lations (solid lines) are compared with experimental
data for serum T, DHT, prostatic AR, prostate weight,
and weight of prostate cell mass and ductal lumen mass
as a fraction of intact values.

biologically plausible behavior and that matches well with events is accurately captured by the model simulations of
available experimental data. postcastration dynamics.
The model captures the kinetics of the endogenous andro- Although cell proliferation, apoptosis, and fluid secretion are
gens and their regulatory effects on the size and function of the each regulated by multiple genes, characterizing each process
prostate in the adult male rat. It also is capable of simulating with a single gene proved to be a reasonable simplification.
intact steady-state conditions and accurately predicting the These simplified empirical descriptions of known complex
experimentally observed sequence of events that occurs after biological processes can be augmented if necessary to include
castration, including the immediate drop in serum androgen greater mechanistic details. Such details may then be incorpo-
concentrations. rated into the model as needed, based on the particular problem
The model also captures the cascade of events that occur to be solved.
following castration and rapid elimination of circulating T This model has a description of the central axis feedback
(except for continuing low levels of adrenal production). The between LH and T that revises that used in Barton and
hypothesis is incorporated that the binding affinities of T and Andersen (6). The new description returns to the same steady
DHT for AR are similar to what is reported in Ref. 53, with state for serum concentrations of T in intact animals after
equilibrium dissociation constants of 0.49 and 0.34 nM, re- perturbations such as an intravenous dose of T or an implant
spectively, but rapid degradation of free AR is prevented by the releasing constant levels of T at physiological or lower con-
two androgens to different extents due to the approximately centrations (simulations not shown). The previous model
threefold faster on and off rates for T than for DHT (53, 54). would establish a new different steady state for serum T
Thus the greater potency of DHT for gene activation in this concentrations. Experimental data are not entirely consistent
model in part reflects greater stability of the receptor ligand on this issue, although return to control steady-state T levels in
complex. This stability initially mitigates the rapid decline in intact animals with implants giving physiological or lower
serum T (occurring in hours), so that prostatic AR declines serum T concentrations is reported by Darras et al. (11),
over days. The consequences of declining AR show up first in Perheentupa et al. (38), and Zirkin et al. (56), supporting the
diminished gene occupancy related to 5␣-reductase activity, new empirical description of the feedback mechanism.
production of prostatic fluid, and finally in decreased cell In a case where limited data are available to qualitatively or
proliferation and increased apoptosis (40). This sequence of quantitatively capture the underlying biology of the central axis

Fig. 11. Model validation. Model simulations (solid lines) are compared with experimental data for prostate weight (left) and prostatic blood flow (right)
following castration as a fraction of intact values. Data shown here were not included in the parameter estimation process.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
E962 MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION

feedback, the model can serve as a tool to test different the intact and castrate steady-state conditions, as well as the
hypotheses about biological mechanisms. Current efforts are dynamics of androgens and the regression of the prostate
underway to explore the dynamics of androgens and prostate following castration. Model simulations match well with mul-
mass in response to external androgen implants. Model simu- tiple types of experimental measurements on both the systemic
lations may be able to motivate new experiments to help and tissue-specific levels, including hormone and AR concen-
elucidate the regulatory mechanisms of androgen levels and trations, prostatic blood flow, enzymatic activity of 5␣-reduc-
AR-mediated prostate maintenance under different hormonal tase, and AR-mediated regulation of prostate size and function.
conditions. Based on these results, the model and the estimated param-
This model has 25 unknown parameters that needed to be eter set seem to provide a plausible representation of the
estimated compared with experimental data. This must be biology. This is further supported by simulations showing that
addressed carefully to minimize problems of mathematical the model matches well with postcastration prostatic blood
ill-posedness and to maximize the biological plausibility of the flow data that were not used in the model calibration process.
model. Parameter estimation problems often are subject to This model validation can be further enhanced as additional
ill-posedness, which means that there may not be a solution to experimental data become available.
the problem, there may be more than one solution, or the By using a systems approach to the gene-regulated processes
problem may be highly sensitive to perturbations in data or involved in prostate maintenance, this model may be used as a
parameters (25). tool for data analysis, generating and testing hypotheses, and
In this case, the unknown parameters first were separated experimental design in the research of the prostate. Moreover,
into two subsets based on function and estimated using two the design of this model can serve as a template for developing
separate estimation problems. As described in METHODS, the models for many other gene-regulated biological processes.
pharmacokinetic parameters were estimated by comparing a The PBPK backbone efficiently describes the transport of
pharmacokinetic submodel to measurements of steady-state relevant compounds through the tissues, while the tissue-
androgen concentrations as in Ref. 6, while the pharmacody- specific pharmacodynamic submodel captures key biological
namic parameters were estimated using the full model com- activity related to protein synthesis and signal transduction.
pared with steady-state concentrations and measurements of The versatility of the model and its capability of capturing
postcastration dynamics. the dynamics of prostate behavior under varying conditions
This approach helped address ill-posedness by splitting the make it useful in both toxicological and pharmaceutical set-
estimation into two problems, effectively reducing the size of tings. Because some pesticides have androgen-antagonistic
the unknown parameter space for each problem. Moreover, the properties (5), augmentation of the model to simulate the
parameters to be estimated were grouped with experimental kinetics and effects of androgen antagonist exposure would
data that were more directly related to the relevant biology for make it a suitable tool for risk assessment. Furthermore, with
each parameter subset (pharmacokinetic vs. pharmacody- enhancements, the model is a potential drug development tool
namic). Incorporating a variety of experimental data also can to predict a response to administration of 5␣-reductase inhib-
help reduce ill-posedness by requiring the model to reproduce itors and androgen antagonists. Both classes of antiandrogens
multiple behaviors. In particular, the postcastration dynamics are frequently used in therapies to treat diseases such as benign
data represent measurements of several different model vari- prostatic hyperplasia and prostate cancer. By calibrating the
ables in time, so that the resulting model simulations capture model with experimental data for a given antiandrogen, the
the biology over several time scales and complexity scales model then can be used to predict the aggregate effect on the
ranging from the receptor level up to the tissue and systemic prostate of similar compounds by adjusting compound-specific
levels. parameters such as binding constants. This type of analysis can
The final step involved solving a third estimation problem yield crucial decision-making estimates of EC50, Emax, and
for all parameters from the first two problems using the full efficacious dose ranges and frequencies, as well as the ability
model. Because the pharmacokinetic parameters were esti- to quantitatively link biomarker data to clinical drug efficacy.
mated using a submodel, it was important to reestimate all The ability of the model to be expanded allows for the
parameters together. The pharmacokinetic submodel did not potential to address a broad range of issues associated with
contain androgen binding to AR, which can significantly alter male reproductive function, especially prostate function and
tissue concentrations by sequestering T and DHT in the pros- maintenance. One major objective is to characterize the effects
tate, for example. The third estimation problem allowed for of prepubertal exposure to antiandrogens, such as vinclozolin,
recalibration of the pharmacokinetic (as well as pharmacody- in male rats, because this has been proposed as a bioassay for
namic) parameters by taking the full model into account. The detecting compounds with endocrine disrupting activity (47).
resulting final parameter values did not significantly change the This will involve adapting the model to describe the normal
model fits to the data for postcastration dynamics but did pubertal maturation process as well as the effects on that
improve the predictions of steady-state androgen tissue con- process that result from prepubertal vinclozolin exposure. Such
centrations. The parameters that changed the most after the an elaboration will require modeling prepubertal regulation
third estimation problem were related to LH synthesis, albu- dominated by androstanediol and the transition to adult andro-
min-androgen binding rates, tissue partition coefficients, and gen regulation dominated by T and DHT. These modeling
the Vmax for 5␣-reductase in the liver. Overall, the final efforts will provide a quantitative means for understanding the
parameter set provides a good fit to the wide variety of data dynamic changes in the male reproductive endocrine system
used in the calibration process. that occur during pubertal development, such as tissue growth,
The results presented here show the ability of the model to androgen production in the testes, and concentrations of an-
reproduce a variety of different biological behaviors, including drogens and binding proteins (2, 3, 13, 14, 34, 44, 47). The
AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION E963
pubertal model will also aid in quantitatively describing how 16. Gatenbeck L, Johansson B, Stromberg L, and Svensson M. Genital
antiandrogens interfere with these pubertal changes, enhance blood flow in male rats subjected to stress stimuli. Urol Res 15: 297–301,
1987.
understanding of the dose-response relationships, and improve 17. George FW. Androgen metabolism in the prostate of the finasteride-
accuracy in the risk assessment of these chemicals (5). treated, adult rat: a possible explanation for the differential action of
testosterone and 5 alpha-dihydrotestosterone during development of the
ACKNOWLEDGMENTS male urogenital tract. Endocrinology 138: 871– 877, 1997.
18. George FW, Russell DW, and Wilson JD. Feed-forward control of
We thank Dr. Roger Rittmaster (GlaxoSmithKline, Research Triangle Park, prostate growth: dihydrotestosterone induces expression of its own bio-
NC) and colleagues at Dalhousie University (Halifax, NS, Canada) for pro- synthetic enzyme, steroid 5 alpha-reductase. Proc Natl Acad Sci USA 88:
viding us with experimental data. 8044 – 8047, 1991.
L. K. Potter’s current address: Scientific Computing & Mathematical 19. Gray LE Jr and Kelce WR. Latent effects of pesticides and toxic
Modeling, GlaxoSmithKline, Research Triangle Park, NC 27709. substances on sexual differentiation of rodents. Toxicol Ind Health 12:
M. G. Zager’s current address: Pharmacokinetics, Dynamics and Metabo- 515–531, 1996.
lism, La Jolla Laboratories, Pfizer, Inc., San Diego, CA 92121. 20. Gray LE Jr and Ostby J. Effects of pesticides and toxic substances on
behavioral and morphological reproductive development: endocrine ver-
GRANTS sus nonendocrine mechanisms. Toxicol Ind Health 14: 159 –184, 1998.
L. K. Potter and M. G. Zager were funded by the Environmental Protection 21. Grino PB, Griffin JE, and Wilson JD. Testosterone at high concentra-
Agency (EPA)/University of North Carolina Toxicology Research Program, tions interacts with the human androgen receptor similarly to dihydrotes-
Training Agreement TA-829472, with the Curriculum in Toxicology, Univer- tosterone. Endocrinology 126: 1165–1172, 1990.
sity of North Carolina, Chapel Hill, NC. This work was reviewed by the EPA 22. Gubbay J, Doyle JP, Skinner M, and Heintz N. Changing patterns of
and approved for publication but does not necessarily reflect official Agency gene expression identify multiple steps during regression of rat prostate in
policy. Mention of trade names or commercial products does not constitute vivo. Endocrinology 139: 2935–2943, 1998.
endorsement or recommendation by the EPA for use. 23. Isaacs JT. Antagonistic effect of androgen on prostatic cell death. Pros-
tate 5: 545–557, 1984.
24. Johansson A, Rudolfsson SH, Wikstrom P, and Bergh A. Altered levels
REFERENCES of angiopoietin 1 and tie 2 are associated with androgen-regulated vascular
1. Andriole G, Bruchovsky N, Chung LW, Matsumoto AM, Rittmaster regression and growth in the ventral prostate in adult mice and rats.
R, Roehrborn C, Russell D, and Tindall D. Dihydrotestosterone and the Endocrinology 146: 3463–3470, 2005.
prostate: the scientific rationale for 5alpha-reductase inhibitors in the 25. Kirsch A. An Introduction to the Mathematical Theory of Inverse Prob-
treatment of benign prostatic hyperplasia. J Urol 172: 1399 –1403, 2004. lems. New York: Springer-Verlag, 1996.
2. Ariyaratne HB and Chamindrani Mendis-Handagama S. Changes in 26. Kyprianou N and Isaacs JT. Activation of programmed cell death in the
the testis interstitium of Sprague Dawley rats from birth to sexual matu- rat ventral prostate after castration. Endocrinology 122: 552–562, 1988.
rity. Biol Reprod 62: 680 – 690, 2000. 27. Lee C. Physiology of castration-induced regression in rat prostate. Prog
3. Ariyaratne HB, Mendis-Handagama SM, and Mason JI. Effects of Clin Biol Res 75A: 145–159, 1981.
tri-iodothyronine on testicular interstitial cells and androgen secretory 28. Lekas E, Johansson M, Widmark A, Bergh A, and Damber JE.
capacity of the prepubertal Rat. Biol Reprod 63: 493–502, 2000. Decrement of blood flow precedes the involution of the ventral prostate in
4. Barton HA and Andersen ME. Dose-response assessment strategies for the rat after castration. Urol Res 25: 309 –314, 1997.
endocrine-active compounds. Regul Toxicol Pharmacol 25: 292–305, 29. Luke MC and Coffey DS. Human androgen receptor binding to the
1997. androgen response element of prostate specific antigen. J Androl 15:
5. Barton HA and Andersen ME. Endocrine active compounds: from 41–51, 1994.
biology to dose response assessment. Crit Rev Toxicol 28: 363– 423, 1998. 30. Maddocks S, Hargreave TB, Reddie K, Fraser HM, Kerr JB, and
6. Barton HA and Andersen ME. A model for pharmacokinetics and Sharpe RM. Intratesticular hormone levels and the route of secretion of
physiological feedback among hormones of the testicular-pituitary axis in hormones from the testis of the rat, guinea pig, monkey and human. Int J
adult male rats: a framework for evaluating effects of endocrine active Androl 16: 272–278, 1993.
compounds. Toxicol Sci 45: 174 –187, 1998. 31. Martel C, Trudel C, Couet J, Labrie C, Belanger A, and Labrie F.
7. Brown RP, Delp MD, Lindstedt SL, Rhomberg LR, and Beliles RP. Blockade of androstenedione-induced stimulation of androgen-sensitive
Physiological parameter values for physiologically based pharmacokinetic parameters in the rat prostate by combination of Flutamide and 4-MA. Mol
models. Toxicol Ind Health 13: 407– 484, 1997. Cell Endocrinol 91: 43– 49, 1993.
8. Charles-River. Research models: products and services 2004. 4-11, 32. Medinsky MA and Klaassen Toxicokinetics CD. In: Casarett and
2004. Doull’s Toxicology: The Basic Science of Poisons (5th ed.), edited by
9. Coyotupa J, Parlow AF, and Kovacic N. Serum testosterone and Casarett LJ, Amdur MO, Klaassen CD, and Doull J. New York: McGraw-
dihydrotestosterone levels following orchiectomy in the adult rat. Endo- Hill, 1996.
crinology 92: 1579 –1581, 1973. 33. Mendel CM. The free hormone hypothesis. Distinction from the free
10. Damassa DA and Gustafson AW. Effects of chronic infusions of sex hormone transport hypothesis. J Androl 13: 107–116, 1992.
steroid-binding protein on the testosterone-mediated inhibition of gonad- 34. Monosson E, Kelce WR, Lambright C, Ostby J, and Gray LE Jr.
otropin secretion and maintenance of sex accessory glands in male rats. Peripubertal exposure to the antiandrogenic fungicide, vinclozolin, delays
Endocrinology 123: 1885–1892, 1988. puberty, inhibits the development of androgen-dependent tissues, and
11. Darras FS, Lee C, Huprikar S, Rademaker AW, and Grayhack JT. alters androgen receptor function in the male rat. Toxicol Ind Health 15:
Evidence for a non-androgenic role of testis and epididymis in androgen- 65–79, 1999.
supported growth of the rat ventral prostate. J Urol 148: 432– 440, 1992. 35. Nnane IP, Kato K, Liu Y, Lu Q, Wang X, Ling YZ, and Brodie A.
12. de Kretser DM, Risbridger GP, and Kerr JB. Basic endocrinology of Effects of some novel inhibitors of C17,20-lyase and 5alpha-reductase in
the testes. In: Endocrinology, edited by DeGroot LJ. Philadelphia, PA: vitro and in vivo and their potential role in the treatment of prostate cancer.
Saunders, 1995, p. 2307–2335. Cancer Res 58: 3826 –3832, 1998.
13. Delp MD, Evans MV, and Duan C. Effects of aging on cardiac output, 36. Omezzine A, Mauduit C, Tabone E, Nabli N, Bouslama A, and
regional blood flow, and body composition in Fischer-344 rats. J Appl Benahmed M. Caspase-3 and -6 expression and activation are targeted by
Physiol 85: 1813–1822, 1998. hormone action in the rat ventral prostate during the apoptotic cell death
14. Diaz E, Castrillon P, Esquifino A, and Diaz B. Effect of prenatal process. Biol Reprod 69: 752–760, 2003.
melatonin on the gonadotropin and prolactin response to the feedback 36a.Ono Y, Suzuki K, Kashiwagi B, Shibata Y, Ito K, Fukabori Y, and
effect of testosterone in male offspring. J Steroid Biochem Mol Biol 72: Yamanaka H. Androgen-dependent blood flow control and morphologi-
61– 69, 2000. cal changes of the capillaries in rat prostate. Int J Androl 27: 50 –56, 2004.
15. Ellison SA and Pardridge WM. Reduction of testosterone availability to 37. Orlowski J, Bird CE, and Clark AF. Androgen metabolism and regu-
5 alpha-reductase by human sex hormone-binding globulin in the rat lation of rat ventral prostate growth and acid phosphatase during sexual
ventral prostate gland in vivo. Prostate 17: 281–291, 1990. maturation. J Endocrinol 116: 81–90, 1988.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.
E964 MATHEMATICAL MODEL FOR ANDROGENIC PROSTATE REGULATION

38. Perheentupa A, de Jong F, and Huhtaniemi I. Biphasic effect of tions Endocrine Disrupter Screening and Testing Advisory Committee.
exogenous testosterone on follicle-stimulating hormone gene expression Crit Rev Toxicol 30: 197–252, 2000.
and synthesis in the male rat. Mol Cell Endocrinol 93: 135–141, 1993. 48. Suzuki K, Ito K, Kurokawa K, Suzuki T, Shimizu N, Fukabori Y,
39. Pozzi P, Bendotti C, Simeoni S, Piccioni F, Guerini V, Marron TU, Honma S, and Yamanaka H. Expression and degradation of rat androgen
Martini L, and Poletti A. Androgen 5-alpha-reductase type 2 is highly receptor following castration, testosterone replacement and antiandrogens
expressed and active in rat spinal cord motor neurones. J Neuroendocrinol administration: analysis by Western blot and immunohistochemistry. To-
15: 882– 887, 2003. hoku J Exp Med 183: 159 –172, 1997.
40. Prins GS. Differential regulation of androgen receptors in the separate rat 49. Tenniswood M, Abrahams P, Winterton V, Bird CE, and Clark AF.
prostate lobes: androgen independent expression in the lateral lobe. J Binding of testosterone, 5 alpha-dihydrotestosterone and 5 alpha-andro-
Steroid Biochem 33: 319 –326, 1989. stane (3 alpha- and 3 beta-), 17 beta-diols to serum proteins in the rat. J
41. Prins GS and Woodham C. Autologous regulation of androgen receptor Steroid Biochem 16: 617– 620, 1982.
messenger ribonucleic acid in the separate lobes of the rat prostate gland. 50. Torres JM and Ortega E. Precise quantitation of 5alpha-reductase type
Biol Reprod 53: 609 – 619, 1995. 1 mRNA by RT-PCR in rat liver and its positive regulation by testosterone
42. Rittmaster RS, Manning AP, Wright AS, Thomas LN, Whitefield S, and dihydrotestosterone. Biochem Biophys Res Commun 308: 469 – 473,
Norman RW, Lazier CB, and Rowden G. Evidence for atrophy and 2003.
51. Vogel CR. Computational Methods for Inverse Problems. Philadelphia,
apoptosis in the ventral prostate of rats given the 5 alpha-reductase
PA: SIAM, 2002.
inhibitor finasteride. Endocrinology 136: 741–748, 1995.
52. Wang JM, McKenna KE, McVary KT, and Lee C. Requirement of
43. Russell DW and Wilson JD. Steroid 5 alpha-reductase: two genes/two
innervation for maintenance of structural and functional integrity in the rat
enzymes. Annu Rev Biochem 63: 25– 61, 1994. prostate. Biol Reprod 44: 1171–1176, 1991.
44. Schoeffner DJ, Warren DA, Muralidara S, Bruckner JV, and Sim- 53. Wilson EM and French FS. Binding properties of androgen receptors:
mons JE. Organ weights and fat volume in rats as a function of strain and Evidence for identical receptors in rat testis, epididymis, and prostate.
age. J Toxicol Environ Health A 56: 449 – 462, 1999. J Biol Chem 251: 5620 –5629, 1976.
45. Shabsigh A, Chang DT, Heitjan DF, Kiss A, Olsson CA, Puchner PJ, 54. Wright AS, Douglas RC, Thomas LN, Lazier CB, and Rittmaster RS.
and Buttyan R. Rapid reduction in blood flow to the rat ventral prostate Androgen-induced regrowth in the castrated rat ventral prostate: role of
gland after castration: preliminary evidence that androgens influence 5alpha-reductase. Endocrinology 140: 4509 – 4515, 1999.
prostate size by regulating blood flow to the prostate gland and prostatic 55. Zhou ZX, Lane MV, Kemppainen JA, French FS, and Wilson EM.
endothelial cell survival. Prostate 36: 201–206, 1998. Specificity of ligand-dependent androgen receptor stabilization: receptor
46. Sharpe RM. Regulation of spermatogenesis. In: The Physiology of Re- domain interactions influence ligand dissociation and receptor stability.
production, edited by Knobil E and Neill JD. New York: Raven, 1994, p. Mol Endocrinol 9: 208 –218, 1995.
1363–1434. 56. Zirkin BR, Santulli R, Awonivi CA, and Ewing LL. Maintenance of
47. Stoker TE, Parks LG, Gray LE, and Cooper RL. Endocrine-disrupting advanced spermatogenic cells in the adult rat testis: Quantitative relation-
chemicals: prepubertal exposures and effects on sexual maturation and ship to testosterone concentration within the testis. Endocrinology 124:
thyroid function in the male rat. A focus on the EDSTAC recommenda- 3043–3049, 1989.

AJP-Endocrinol Metab • VOL 291 • NOVEMBER 2006 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (090.003.152.237) on March 15, 2024.

You might also like