03.12.2012 Views

Premenstrual Syndromes : PMS and PMDD - Rutuja :: The site ...

Premenstrual Syndromes : PMS and PMDD - Rutuja :: The site ...

Premenstrual Syndromes : PMS and PMDD - Rutuja :: The site ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

a substance/enzyme is being measured. <strong>The</strong>se studies<br />

may enable investigators to determine both the metabolic<br />

consequences of ovarian steroid exposure, as well<br />

as the identity of c<strong>and</strong>idate mediators of the differential<br />

behavioral response (i.e. altered pattern of metabolites)<br />

observed in women with <strong>PMDD</strong>.<br />

A second possible <strong>site</strong> of differential steroid signal<br />

modification is at the level of the steroid receptor.<br />

Steroid receptor function is modified by an array of<br />

tissue-specific factors including the relative concentration<br />

of co-regulator proteins, the presence <strong>and</strong> activity<br />

of receptors for other members of the steroid family or<br />

receptor subtypes, <strong>and</strong> concurrent action of the steroid<br />

at the cell membrane. In human disease one of the best<br />

characterized sources of variation in steroid receptor<br />

function is genomic. Polymorphisms in gonadal steroid<br />

receptors have been shown to alter receptor transcriptional<br />

efficacy (e.g. CAG repeat in exon 1 of the <strong>and</strong>rogen<br />

receptor; progins insertion in intron 7 of the progesterone<br />

receptor; T1057G in exon 5 of �-estrogen receptor)<br />

<strong>and</strong> to be associated with differential illness risk<br />

(i.e. prostate cancer, breast cancer). 52–55 Additionally,<br />

the susceptibility to the disruptive effects of estradiol<br />

on reproductive development differs enormously (up to<br />

100-fold) between mouse strains, with the genotype<br />

contributing more to the variance than the dose of<br />

estradiol employed. 56 Finally, a single nucleotide polymorphism<br />

enables progesterone to activate a receptor,<br />

the mineralocorticoid receptor, at which it normally<br />

functions only as a competitive inhibitor, thus providing<br />

a means by which normal steroid levels create a different<br />

phenotype (hypertension). 57 <strong>The</strong>re is precedent, then,<br />

for expecting that polymorphisms in the gonadal steroid<br />

signaling pathway or in gonadal steroid-regulated genes<br />

could alter the nature or strength of the steroid signal<br />

as well as phenotype. Alternatively, non-steroid-related<br />

genes could be implicated in the vulnerability to develop<br />

the differential behavioral response to ovarian steroids<br />

in <strong>PMDD</strong>. For example, Caspi et al 58 demonstrated<br />

that the risk of developing depression is predicted by<br />

the experience of stressful life events in combination<br />

with a specific genetic background but is not predicted<br />

by either the genetic or environmental factors when<br />

considered separately. Thus gene–environment interactions<br />

represent a promising explanation for the differential<br />

behavioral sensitivity to gonadal steroids seen in<br />

women with <strong>PMDD</strong>. Analogous to the suggestion by<br />

Caspi et al, <strong>PMDD</strong> symptoms could develop after a<br />

hormonal stimulus (i.e. the environmental event)<br />

against the background of a specific genetic context.<br />

Whereas some earlier c<strong>and</strong>idate gene studies did not<br />

find significant associations with <strong>PMDD</strong>, 59 we have<br />

recently identified a region of the ESR1 gene containing<br />

multiple polymorphic alleles that associate with <strong>PMDD</strong>,<br />

thus lending support to the idea that the effects of multiple<br />

genes may interact in creating a dysphoric behavioral<br />

response to normal gonadal steroid levels. 64<br />

<strong>The</strong> contributions of genotypic variation to <strong>PMDD</strong><br />

phenotype could be modified by several other factors,<br />

including both epigenetic processes as well as gene–<br />

environment or gene–steroid interactions. For<br />

example, Meaney et al 60,61 have demonstrated that the<br />

behavioral phenotype can be determined by environment-induced<br />

alterations in the expression of the<br />

genome. Thus, early-life trauma or past episodes of<br />

depression, both frequent accompaniments of<br />

<strong>PMDD</strong>, 62 could potentially modify the substrate<br />

response to the gonadal steroid trigger. As more is<br />

learned about both the nature of gonadal steroid<br />

signaling in the brain <strong>and</strong> the complexities of gonadal<br />

steroid-regulated behaviors, the sources of the differential<br />

response to gonadal steroids in <strong>PMDD</strong> will be<br />

clarified. In summary, women with <strong>PMDD</strong> could have<br />

excessive or deficient gonadal steroid signaling that<br />

might alter the processing of stressors <strong>and</strong> lead to a<br />

dysregulated affective response 63 or altered learning<br />

that could favor the development of behavioral sensitization<br />

or steroid-dependent interoceptive cueing of<br />

behavioral states.<br />

CONCLUSIONS<br />

In <strong>PMDD</strong> there is a well-defined endocrine stimulus<br />

that for many women with this condition is critical in<br />

the precipitation of affective state disturbances. This<br />

affords a unique opportunity to identify neural substrates<br />

involved in the regulation of the affective state<br />

that could be targeted in the development of novel<br />

treatment strategies. Our pursuits of several testable<br />

hypotheses in <strong>PMDD</strong> will advance our underst<strong>and</strong>ing<br />

of this disorder, illuminate mechanisms by which reproductive<br />

steroids may regulate the affective state, <strong>and</strong><br />

help identify the locus of the differential sensitivity that<br />

permits reproductive steroids to destabilize mood in<br />

some but not all women. A better underst<strong>and</strong>ing of the<br />

pathophysiology of this complex disorder will permit the<br />

development of theoretically driven treatment strategies<br />

<strong>and</strong>, hopefully, predictors of therapeutic response<br />

for the considerable number of women with <strong>PMDD</strong><br />

who are not responsive to either ovarian suppression<br />

strategies or SSRIs.<br />

ACKNOWLEDGMENT<br />

FUTURE TREATMENTS 177<br />

<strong>The</strong> research was supported by the Intramural Research<br />

Programs of the NIMH.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!