Neurologic Disease in Women - part 3 pdf

50 555 0
Neurologic Disease in Women - part 3 pdf

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

NEUROLOGIC DISEASE IN WOMEN 82 Progesterone genomically influences the enzymatic activ- ity controlling the synthesis and release of various neu- rotransmitters and neuromodulators (7). Progesterone decreases the number of dendritic spines and synapses on hippocampal CA1 pyramidal neurons, thus counter- acting the stimulatory effects of E2 (33). It has inhibitory direct membrane effects, described in the next section. Neuroactive Steroids The anticonvulsant effect of progesterone is largely medi- ated by its 3 ␣-hydroxylated metabolite, 3-␣-hydroxy-5-␣- pregnan-20-one or allopregnanolone (AP) (37,38). Allo- pregnanolone and the 3␣,5␣-hydroxylated natural metabolite of the mineralocorticoid deoxycorticosterone, allotetrahydro-deoxycorticosterone (allo-THDOC), are the two most potent of a number of endogenous neu- roactive steroids with a direct membrane effect on neu- ronal excitability (37,39). Allopregnanolone is devoid of hormonal effects. It may be thought of as an endogenous regulator of brain excitability with anxiolytic, anticon- vulsant, and sedative-hypnotic properties (37). Allopreg- nanolone and allo-THDOC hyperpolarize hippocampal and other neurons by potentiating GABA-mediated synap- tic inhibition. They act as positive allosteric modulators of the GABA-A receptor, interacting with a steroid-specific site near the receptor to facilitate chloride (Cl) channel opening and prolong the inhibitory action of GABA on neurons (7,37,39). Allopregnanolone is one of the most potent ligands of GABA-A receptors in the CNS, with affinities similar to the potent benzodiazepines and approximately a thousand times higher than pentobarbi- tal (37,39). Progesterone by itself enhances GABA-induced Cl- currents only weakly and only in high concentrations (37). Plasma and brain levels of allopregnanolone paral- lel those of progesterone, and plasma levels of AP corre- late with progesterone levels during the menstrual cycle and pregnancy (37). Brain activity of progesterone and AP, however, is not dependent solely on ovarian and adrenal production, because they are both synthesized de novo in the brain (40). Their synthesis is region-specific and includes the cortex and the hippocampus. Allopregnanolone and allo-THDOC have potent anticonvulsant effects in animal seizure models and in sta- tus epilepticus (37,38,41). Allopregnanolone’s anticon- vulsant properties resemble those of clonazepam, but with lower relative toxicity and with little habituation to its anticonvulsant effect (42). The abrupt withdrawal of allo- pregnanolone induces seizures, possibly by a modulation of the ␣-4 GABA-A receptor subunit that confers GABA- insensitivity on the GABA-A receptor. This may be a mechanism of the perimenstrual seizure exacerbation seen in some women with epilepsy (43). Although the 3- and 5-␣-reduced steroids potentiate GABA-A receptor activity and enhance neuronal inhibi- tion, some of the sulfated neuroactive steroids have neu- roexcitatory effects. These include pregnenolone sulfate and DHEAS, the naturally occurring sulfated esters of the progesterone precursor pregnenolone and of the proges- terone metabolite DHEA (Figure 6.4; 37,44). These steroids increase neuronal firing when directly applied to neurons by antagonizing GABA action at the GABA-A receptor and by facilitating glutamate-induced excitation at the NMDA receptor (45). In animals, pregnenolone sulfate and DHEAS have a proconvulsant effect that is prevented by chronic pretreatment with progesterone (37,45). These neurosteroids may also affect cognition and memory (46). Pregnenolone sulfate stimulates ACh release as well as glutamatergic activity in adult rat hipp- pocampus. DHEAS and PS improve memory and learn- ing in aging mice. In humans, DHEA has been reported to have mood elevating and memory-enhancing effects in middle-aged healthy men and women and in patients with depression (47,48). Trophic Effects In addition to their neuromodulatory effect on neuronal excitability, gonadal steroids also exert a trophic effect on neurons and glial cells. Estrogen is an important neural trophic factor throughout life, with influences on neuronal differentiation, survival, and plasticity (49–51). The trophic effects of estrogen may ameliorate the degeneration of neurons in diseases such as Alzheimer disease (see also Chapter 30), reduce apoptotic neuronal death in ischemic and traumatic brain injury, and pro- mote neuronal regeneration and growth following such injuries. Trophic Effects and Cholinergic Function in Basal Forebrain and Cortex Estrogen plays an important role in the function of the basal forebrain cholinergic system involved in memory and cognition. In ovariectomized rats, estradiol replace- ment improves spatial memory and maze learning (52). The basal forebrain cholinergic neurons of the nucleus basalis myenert (NBM) and of the diagonal band of Broca (DBB) innervate the forebrain and the hippocampus, areas important in cognition, learning, and memory. Their degeneration is a key feature of Alzheimer disease. Estradiol protects cholinergic neurons against exci- totoxic neuronal damage (53). It does so by potentiating the endogenous trophic effects of the neurotrophins, nerve growth factor (NGF), and brain-derived neu- rotrophic factor (BDNF). These trophins are produced in the target areas of basal forebrain cholinergic projec- tions and exert a trophic effect on cholinergic neurons by binding with specific tyrosine kinase receptors, tyro- EFFECT OF OVARIAN HORMONES ON THE NERVOUS SYSTEM 83 sine kinases A and B (trkA and trkB). TrkA and BDNF mRNA levels in the cholinergic neurons fluctuate across the estrous cycle in parallel with estrogen levels (54). Estradiol and estradiol with progesterone increase trkA and trkB levels in the basal forebrain cholinergic neurons and BDNF in the hippocampus (55). The protective effects of estrogen on cholinergic neurons may underlie the observed protective effect of postmenopausal estro- gen replacement therapy against the development of Alzheimer disease (26,56,57). Role in Neuronal Injury and Neuroprotection Estradiol protects neurons against a wide variety of neu- rotoxic stimuli, including ischemic CNS injury, oxidative stress, excitotoxic insults, and b-amyloid-induced toxic- ity (49,50,58). In the middle cerebral artery (MCA) occlusion animal model of cerebrovascular accident (CVA), low levels of estradiol replacement reduce infarct size by 50%. The treat- ment must precede ischemia by several days (59). Low-dose estradiol pretreatment has a similar neuroprotective effect on the pyramidal CA1 neurons of the hippocampus in sta- tus epilepticus in rats (60) and protects explant cultures of both neurons and astrocytes against cell death. The mechanism of this neuroprotection may include the estrogen receptor–mediated inhibition of the apop- totic signaling pathways (58), regulation of growth fac- tor genes and their receptors, and modulation of neurite outgrowth and plasticity (51). In the neocortex, estrogen ␣ receptor (ER-␣) is expressed at high levels only during development, when neocortical differentiation occurs, thus suggesting a developmental role (10). Neocortical estrogen ␤ receptor (ER-␤), by contrast, is expressed throughout life. In adulthood, ER-␣ is expressed in the cortex only after neuronal injury such as CVA. In ER-␣ knockout rats, estradiol has no protective effect against CVA (58). Thus, ischemia or injury induces the expres- sion of ER- ␣, the activation of which by estradiol protects against ensuing neuronal injury. Recently, another mem- brane-associated estrogen receptor (ER-X) has been iden- tified; ER-X is also expressed perinatally and is only expressed in adulthood following neuronal injury such as stroke. Its activation may also be involved in injury- related neuroprotection (49–51). Pregnenolone also may be important in neuropro- tection. It reduces the degree of the histopathological injury and increases the recovery of motor function in rats after traumatic spinal cord injury (61). The mechanism is unclear. Other poorly understood, potentially neuroprotec- tive effects include a reduction of cerebral edema by prog- esterone following cortical contusion, first suggested by the observations that males have more edema after simi- lar degrees of cortical contusion than females (61). Myelination Finally, gonadal steroids may play a hitherto little appre- ciated role in myelination. Oligodendrocytes and Schwann cells express estro- gen and progesterone receptors. Estradiol increases the proliferation of Schwann cells. This finding may relate to the exacerbation of neurofibromatosis during perime- narche (62). Schwann cell synthesize progesterone from preg- nenolone. Progesterone synthesis may be important in myelin formation. Expression of the synthesizing enzyme, 3- ␤-hydroxysteroid dehydrogenase (3␤HSD) and prog- esterone synthesis increase in Schwann cells during myelin formation. Progesterone, in turn, promotes myelin for- mation by Schwann cells. Following cryolesion of the sci- atic nerve, progesterone concentrations in the regenerat- ing nerve are about sixfold higher than in plasma. Blocking progesterone synthesis or receptor inhibits the formation of new myelin. Conversely, local application of progesterone or pregnenolone accelerates remyelina- tion (61,63). Oligodendrocytes also express progesterone recep- tors and 3␤HSD, and progesterone may also promote myelination in the CNS. CLINICAL IMPLICATIONS Genetically Based Disorders Disorders that have a genetic basis may encompass an altered ovarian hormonal production, which may affect neurologic function and may affect those neurologic dis- orders that have a recognized relationship to fluctuations in cyclical hormones. Most of these disorders are dealt with in a more detailed fashion in other chapters of this book, but a few of these conditions deserve additional comments here. Turner syndrome is an example of a chromosomal deletion. About 1 in every 5,000 live-born females has 45 chromosomes plus a single X chromosome; that is, there is a deletion of one X chromosome. Girls have ovarian dysgenesis, absence of ovarian hormonal secretion, high FSH levels, and delayed adolescence as well as a number of associated somatic developmental anomalies. When sexual maturation is desired, patients must be treated with exogenous hormone replacement. Women with Turner syndrome exhibit male cognitive patterns—they perform better on visuospatial tasks than on verbal tasks. When untreated with estrogen, patients with Turner syn- drome have memory, attention, and spatial performance impairment and hippocampal volume loss on magnetic resonance imaging (MRI) (64). Another genetically based disorder is congenital adrenal hyperplasia (CAH). This autosomal recessive dis- NEUROLOGIC DISEASE IN WOMEN 84 order can be caused by a defect in one of six recognized steroid synthesizing enzymes. It affects both men and women. In three forms, only the adrenal gland is affected. In the other three, both the adrenal gland and the ovary are affected. The enzymatic deficiency (e.g., of the CYP450-c21 hydroxylase) results in impaired adrenal synthesis of cortisol, reduced inhibitory feedback of ACTH, and increased adrenal synthesis of the cortisol precursors that can be converted to androgens. Clinically, women with this condition have mild to moderate viril- ization that manifests itself early in life, and that is occa- sionally associated with a delay in the onset of sexual development. Two clinical forms of the disease present neonatally, one in late childhood, adolescence, or adult- hood. In the neonatal forms, there is an increased prena- tal production of androgens. The classic form of CAH due to 21-hydroxylase deficiency is a rare disorder of adrenal steroid synthesis that affects approximately 1 in 15,000 live births as a result of a gene mutation on the short arm of chromosome 6. Males or females with CAH are exposed to high levels of androgens during gestation, beginning in the third month of fetal life. As the disease is now readily diagnosable and treatable at birth, the hor- monal abnormalities are confined to prenatal and early neonatal exposure. CAH has been associated with some behavioral changes that have been attributed to intrauter- ine exposure to increased androgen levels. Women with CAH have an increased risk of gender identity disorder (e.g., of adopting male sexual identity), increased inci- dence (33%–45%) of homosexual tendencies, and show masculine play behavior in childhood and male-typical cognitive performance in adulthood (65). In addition, women with CAH have a higher incidence of polycystic ovarian syndrome, which may have neurologic conse- quence (2). Physiologic Disorders Changes in the secretion of ovarian hormones associated with menarche, menstrual cycles, pregnancy, and menopause may all affect the clinical manifestation of a number of disorders such as epilepsy, migraines, multi- ple sclerosis, movement disorders, and pseudotumor cere- bri during a woman’s life. Partial Epilepsy Several researchers have noted that epilepsy commonly starts around the time of menarche (66,67). In one study, seizures began at menarche in 19% of all adult women with epilepsy. In another study, 35% of epilepsy that began between the ages of 0.5 and 18 years began within 2 years of menarche. Epilepsy was much more likely to start within 2 years of menarche (perimenarche) and dur- ing the year of menarche than during any other postna- tal childhood period (66). In girls with pre-existing epilepsy, approximately one-third experience seizure exacerbation during puberty (66–68). This is more likely to occur in girls with focal epilepsies, refractory seizures, evidence of CNS damage, and delayed menarche. Changes in reproductive hormones may be respon- sible for these observations. Sexual maturation begins with adrenarche, which starts between the age of 8 and 10 with a marked increase in the secretion of DHEAS and DHEA (69). This is followed by gonadarche, which starts around the age of 10 with the secretion of estrogen, but without the secretion of progesterone. The ovarian secre- tion of estrogens gradually rises through menarche (median age 12.8 years) until the onset of ovulation. In the majority of girls, menstrual cycles are initially anovu- latory. Ovulation only starts 12 to 18 months after menar- che. It is only at this point that the ovarian secretion of progesterone begins, with a parallel increase in serum allopregnanolone levels in late puberty (70). Thus, the secretion of the neuroexcitatory steroids, DHEAS and estrogen, precedes the secretion of progesterone, the neu- roinhibitory steroid, by several years. Continued expo- sure of the brain during this time to the proconvulsant effects of estrogen and DHEAS without the anticonvul- sant effect of progesterone may facilitate the development of epilepsy (epileptogenesis) in susceptible girls. The cyclical pattern of estradiol and progesterone secretion may influence the likelihood of seizures (36). Catamenial seizures broadly refer to an identifiable and predictable occurrence of seizures in relationship to the menstrual cycle (28,71–73). Herzog et al. described three patterns of catamenial seizure exacerbation (74). The two more easily recognized patterns are (i) worsening of seizures during the mid-cycle and (ii) perimenstrually in women with normal ovulation. In the first case, the occur- rences of seizures coincide with ovulation, whereas in the second form, the occurrences happen 1 to 2 days before the onset and 1 to 2 days after the onset of menstruation. The third pattern occurs in women who fail to ovulate, when seizures occur throughout the entire late stage of the cycle, which may vary considerably in duration. It is sometimes easier to note that seizures decrease in occur- rence from day 2 through days 8 to 10, and then increase until menstruation. As mentioned earlier, estradiol has proconvulsant effects on the brain, whereas progesterone has anticon- vulsant effects. In women with ovulatory cycles, the surge of ovarian secretion of estrogen before and during ovu- lation may be responsible for the periovulatory seizure exacerbation. During the luteal phase, the anticonvulsant effect of progesterone secreted by the corpus luteum may protect against seizures, resulting in lower seizure fre- quency (71,72,74). Perimenstrual seizure exacerbation may be due to the withdrawal of progesterone and its GABA-mediated anticonvulsant effect, similar to the EFFECT OF OVARIAN HORMONES ON THE NERVOUS SYSTEM 85 withdrawal seizures seen with a discontinuation of bar- biturates, benzodiazepines, or alcohol (42,43). In women with anovulatory cycles, the ovary secretes essentially normal quantities of estrogen during the late follicular and luteal phases (not the periovulatory phase) but does not secrete progesterone. Thus, an elevated estrogen:prog- esterone ratio occurs from late follicular phase until men- struation. This may explain the unusual pattern of seizure exacerbation, when seizures occur from about menstrual cycle day 8 to 10 until menstruation. In essence, such women are only protected against seizure exacerbation when the ovary secretes very little estrogen during the early and mid-follicular phase of the cycle. Menopause may also affect epilepsy. The term menopause refers to a complex process that encompasses both menopause, cessation of all menstruation, and per- imenopause, the preceding decline in reproductive endocrine function. Perimenopause often extends for sev- eral years. Early in perimenopause ovulatory cycles change to anovulatory, and progesterone secretion declines (75). By contrast, estrogen secretion remains nor- mal through most of perimenopause and may even increase episodically when, as a result of erratic follicu- lar development, multiple follicles develop during some menstrual cycles. Estrogen levels only drop consistently late in the perimenopause, during the last few months before cessation of menses, as the follicle pool becomes exhausted. Thus, for a period of time that may last for several years, there may be a relative excess ratio of estro- gen to progesterone. Based on the pattern of hormonal change, an evolving seizure pattern with seizure exacer- bation during the perimenopause might be expected: ini- tial seizure exacerbation when progesterone secretion declines but estrogen secretion continues, followed by sta- bilization or improvement after menopause, as estrogen secretion ceases. This pattern did, in fact, occur in a recent study (76). Sixty-four percent of women experienced seizure exacerbation, and only 13% of women experi- enced seizure improvement during the perimenopause. By contrast, 43% of women had seizure improvement dur- ing the menopause, with only 31% experiencing seizure exacerbation. Partial epilepsy may also begin during the climacteric, sometime without an apparent cause (77). It is possible that the chronic exposure of the brain to estro- gen without progesterone during the perimenopausal years could “kindle” an occult nonepileptic CNS lesion into an epileptic one, in a way similar to the suggested epileptogenic effect of perimenarche. Estrogen replace- ment therapy may also be associated with seizure exac- erbation during the perimenopause and menopause (76). We believe that if there is a clinically significant increase in seizure frequency, hormonal replacement should include both estrogen together with natural progesterone. In addition, epilepsy, particularly temporal lobe epilepsy, can influence the menstrual cycle. As mentioned, the amygdala, a mesial temporal lobe structure, has reciprocal relationships with hypothalamic structures that influence gonadotrophin secretion. In our study of 50 women with clinical and electroencephalographic evi- dence of temporal lobe onset partial epilepsy, 38% had significant reproductive abnormality (78). Approximately 20% had polycystic ovarian syndrome (PCOS), and 12% had hypogonadotrophic hypogonadism (HH). Two of the women had premature menopause, and one had hyper- prolactinemia. An increased risk of premature menopause among women with epilepsy was also observed in another study (79). In humans, it appears that a significant right temporal lobe versus left temporal lobe differential effect occurs in the hypothalamic gonadotrophin response to temporal lobe seizure activity. We first observed that the LH levels in women with temporal lobe epilepsy varied considerably compared to age-matched controls (80). Women with left temporal seizures had more LH surges during an 8-hour period than controls. These women all had PCOS. In women with hypothalamic hypogonadism (HH), there was a marked decrease in the number of LH surges during an 8-hour period compared to controls, and the seizure focus was more often right-sided. A possible explanation for these findings may include a differential effect of altered input from the right and left amygdala on the hypothalamic GnRH neuronal pulsatile activity (80). In addition to the above observations regarding the complex interactions of seizure type and seizure location on hormonal cyclicity and the hormonal effect on seizure frequency, medications play an important and often con- founding role. Similarly, pregnancy may have a major effect on seizures through its effect on endogenous hor- mone production and its effect on the metabolism of the antiseizure medication. These effects are discussed in more detail in a later chapter. Migraine Migraine is equally prevalent in boys and girls until ado- lescence, when the ratio changes to 3:1 in favor of women: 17.6% of women suffer from migraines com- pared with 5.7% of males (81). In approximately 60% of women, migraine attacks are linked to the menses, and in approximately 15% of women with migraines, attacks occur exclusively perimenstrually. The catamenial exac- erbation of migraines begins at menarche in approxi- mately 33% of women with menstrual migraines. During pregnancy, migraines may worsen during the first trimester and remit during the last two trimesters, although the pattern of improvement or exacerbation is highly variable and individual; approximately 25% of women with migraines experience no change in their headaches during pregnancy (82). Migraines may worsen transiently, but at times markedly and for a prolonged time, during perimenopause; migraines may improve after NEUROLOGIC DISEASE IN WOMEN 86 completion of menopause when the female:male ratio drops to 2:1 (83). The pathophysiologic underpinnings of these clini- cal phenomena remain essentially obscure. A popular hypothesis is that estrogen withdrawal perimenstrually alters vascular tone, leading to vascular instability and a greater susceptibility to cerebrovascular dilatation and headache. Estrogen receptors are found on the media of medium-size cerebral vessels. Estrogen stimulates the pro- duction of nitric oxide and causes cerebrovascular dilata- tion (84). Blood flow in the internal carotid artery increases by 15% during the ovulatory phase of the men- strual cycle in normal women (85). However, no differ- ence has been found in the systemic levels of estrogens, progesterone, androgens, LH, or FSH between women with catamenial migraines and controls (86). No blood hormone–blood flow correlation studies have been per- formed in women with migraines. Progesterone has not been thought to be a significant factor in migraine, but it is noteworthy that in an animal model of migraine, pre- treatment with both progesterone and the 3,5-a reduced metabolites allopregnanolone and tetrahydrodeoxycorti- costerone ameliorated plasma extravasation within the meninges (87). This would suggest that progesterone— via allopregnanolone—may play an anti-inflammatory role in the CNS. Perimenstrual withdrawal of proges- terone could thus theoretically contribute to an increase in the vasogenic inflammation that may be part of the pathophysiology of migraine. Other possible mechanisms that have been sug- gested include a perimenstrual reduction of hypothala- mic opioid secretion, increased prostacyclin activity, and prostacyclin-related vasodilation and modulation of pro- lactin secretion (83). Of particular interest is the influ- ence of estrogen on opioids. Estradiol colocalizes with the opioids endorphins, encephalin, and dynorphin in rat neurons of a number of brain regions, including the hypothalamus and the dorsal spinal cord sensory neu- rons. It induces the expression and release of the endoge- nous opioid peptides and activate µ-pioid receptor acti- vation in the hypothalamus and in the amygdala (88). Expression of endorphin in hypothalamic neurons and the release of opioids into the hypothalamic-portal cir- culation fluctuates during the menstrual cycle. It is high- est at the time of ovulation (estrus) and falls as serum estrogen levels fall (89). Thus, estradiol potentiates the analgesic effects of endogenous opioids. It may, possibly, by its effect in the amygdala, even alter the subjective perception or “emotional content” of painful stimuli. Its withdrawal perimenstrually may contribute to the men- strually related migraine. Conversely, its large rise dur- ing the last two trimesters is associated with an elevation of the pain threshold during gestation (90). Thus, it may contribute to the alleviation of migraine during this part of pregnancy. These theories have led to limited therapeutic trials with estrogen and, paradoxically, antiestrogen therapy, for example, with tamoxifen, with androgens such as danazol, and with dopamine agonists such as bromocrip- tine and pergolide to suppress prolactin secretion (91). These studies have been limited in scope and therapeutic success, although anecdotal reports of success using all these agents abound. Multiple Sclerosis Multiple sclerosis (MS) is also more common in women than men, with approximately a 2.5:1 ratio. The onset of the disease is also most common during the second and third decades of life, although the incidence rises after puberty, during the second half of the second decade. Anecdotal reports of MS show perimenstrual exacerba- tion (92), but also improvement with estrogen contra- ceptive treatment (93). One of the most notable features of MS, however, is the reduction of relapsing attacks in remitting and relapsing MS during the last trimester of pregnancy, with a subsequent rebound of attacks during the postpartum period (94). The relapse decrease of the last trimester may be mediated by a shift in immune responses from the inflam- matory response promoting T helper 1 lymphocytes (Th1 cells) to the inflammatory response dampening T helper 2 lymphocytes (Th 2 cells). A number of hormones rise dramatically during the second half of pregnancy. The serum levels of estradiol, estriol, progesterone, cortisol, and 1,25-vitamin D, among others, rise tenfold during this time, compared with their preconception levels. All these hormones affect the immune system. Estradiol, estriol, cortisol, and 1,25-vitamin D have been shown to have an immunosupressant effect and a suppressant effect on experimental allergic encephalomyelitis (EAE), the ani- mal model of MS (95). Estrogens affect CD4+ T lym- phocytes, with differential effects at low versus high dose. High levels of estrogen favor T-2 anti-inflammatory cytokine and humoral immune response (96). Proges- terone also facilitates the T-2 profile, with the induction of the messenger RNA of the anti-inflammatory inter- leukin-4 (97). Clinically, the number and volume of gadolinium- enhancing MRI lesions in women with MS do not fluc- tuate between the follicular and the luteal phases of the menstrual cycle. A positive relationship, however, has been demonstrated between MRI lesion number and vol- ume and the serum progesterone:estradiol ratio (98). Attempts at the therapeutic manipulation of repro- ductive hormones other than in MS have not been system- atic and have been largely unsuccessful. Bromocriptine, which suppresses the secretion of prolactin, was found to be very effective in suppressing EAE in animals when administered both before and after the EAE-inducing agent EFFECT OF OVARIAN HORMONES ON THE NERVOUS SYSTEM 87 (95). Attempts at human studies, however, were not promising and have been abandoned (99). 1,25-vitamin D was similarly promising in EAE models and disappointing in limited human studies (100). Recently, the weak estro- gen estriol, a major estrogen product of the second half of human pregnancy, was found to suppress EAE and to decrease delayed-type hypersensitivity responses in periph- eral blood mononuclear cells and gadolinium-enhancing MRI lesion number and volumes in nonpregnant women with MS compared with pretreatment baseline. The bene- ficial MRI effects receded when the treatment was stopped and re-emerged when it was reinstituted (101). A placebo- controlled study is being planned. Neuropsychiatric Diseases As already mentioned, most neuropsychiatric diseases are “sexually dimorphic,” with a greater predilection for women (depression, anxiety disorders, anorexia-bulimia) or for men (aggression, schizophrenia) (16). The differ- ences in incidence and prevalence of these disorders between men and women emerges during puberty. Menar- che has been aptly named “the forgotten milestone” of female psychiatric diseases (16). Affective and anxiety dis- orders are commonly affected by the menstrual cycle, and commonly exacerbate or present de novo during the post- partum period or during the perimenopause (22). Both estrogens and progesterone have psychoactive properties. Estrogens, via diverse mechanisms that may include augmentation of NMDA and non-NMDA gluta- matergic activity, serotonergic, noradrenergic, and opiate activity, have an arousing, antidepressant, and potentially anxiogenic effect (102). Progesterone and allopreg- nanolone, by contrast, have anxiolytic, sedating and, in higher doses, depressive and anesthetic effects similar to those of the benzodiazepines, due to their potentiation of GABA-ergic activity. Progesterone withdrawal may there- fore be pathophysiologically important in the perimen- strual exacerbation of anxiety disorders, and of rapid cycling in bipolar affective disorders, and in premenstrual dysphoric dysfunction (PMDD) or premenstrual syndrome (PMS). PMDD women with greater levels of premenstrual anxiety and irritability have significantly reduced allo- pregnanolone levels in the luteal phase relative to less symptomatic PMDD women (103). This suggests that a dysfunction of metabolism of progesterone to allopreg- nanolone may be one factor in the causation of PMDD. The withdrawal of progesterone and low serum allopreg- nanolone levels may also be implicated in postpartum depression. Serum allopregnanolone levels were similarly decreased after delivery in women with postpartum dys- thymia compared to euhymic women (104), with a nega- tive correlation between Hamilton Depression Rating score and serum allopregnanolone level. A significant neg- ative correlation was observed between the Hamilton score and levels of serum allopregnanolone. Movement Disorders Parkinsonian symptoms can worsen perimenstrually in women with Parkinson disease. In one large survey, 75% of women with natural menstrual cycles noted a wors- ening of symptoms before or during the period (105). The pathophysiologic mechanisms have not been investigated. In chorea gravidarum, chorea occurs during preg- nancy, sometimes in patients with previous post- rheumatic fever chorea (Sydenham chorea). Its patho- genesis is unclear, but may be related to a pregnancy-associated rise in gonadal hormones, partic- ularly estrogens. This hypothesis is supported by the observation that estrogen-containing oral contraceptive may be a trigger for chorea, sometimes in a patient who also suffers from chorea gravidarum (106). (See also Chapter 24.) CONCLUSION The study of the effects of hormones on the nervous sys- tem, mood, memory, cognition, and behavior in health and in disease is beginning to receive the attention that it deserves. Hopefully, over the next few years, the complex interrelationships between hormonal fluctuations and the various neurotransmitter systems and metabolic path- ways, as well as neuronal survival, brain plasticity, neu- ronal remodeling, and synaptogenesis will be more fully understood so that we might predict and treat the normal and pathologic conditions that arise from the cyclical behavior of ovarian hormones. On a final note, a word of caution. Although a good deal is known about the effects of ovarian hormones on the nervous system, very little is known about two aspects that may be important. The first is the adaptive response of the nervous system to the fluctuation levels of the steroids. Serum steroid levels may change dramatically without clinical effects. During the last trimester of the pregnancy, for instance, serum levels of progesterone and estradiol rise to approximately 10 times the level of the luteal phase of the menstrual cycle and approximately 40 to 200 times the level of the early follicular phase. Within 24 to 48 hours after delivery, the secretion returns to the follicular phase level. Yet in the majority of women, no neurologic complications occur during the last trimester of the pregnancy or the puerperium (2). Thus, adaptive changes must mitigate the effects of such large fluctuations in serum levels on the nervous system. Second, we know very little about the functional sig- nificance of in situ synthesis of neurosteroids in the CNS. This synthesis is larger than peripheral steroid synthesis for several major gonadal and adrenal steroids such NEUROLOGIC DISEASE IN WOMEN 88 DHEA, DHEAS, and pregnenolone (brain levels of which are up to 10 times higher than serum levels), as well as for neuroactive progesterone metabolites such as allopreg- nanolone and TH-DOC (105). Such knowledge will be important in determining the overall role of steroids, including ovarian steroids, in the healthy and diseased functioning of the nervous system. References 1. Greenspan FS, Strewler GJ, (eds.) Basic and clinical endocrinology , 5th ed. Stamford, Conn: Appleton and Lange, 1997. 2. Yen SSC, Jaffe RB, Barbieri RL, (eds.) Reproductive endocrinology , 4th ed. Philadelphia, Pa: WB Saunders, 1999. 3. Funabashi R, Shinohara K, Kimura F. Neuronal control circuit for the gonadotropin-releasing hormone surge in rats. In: Handa RJ, Hayashi S, Terasawa E, Kawata M, (eds.) Neuroplasticity, development and steroid hormone action . Boca Raton, Fla: CRC Press, 2002;169–177. 4. Dreifuss JJ, Murphy JT, Gloor P. Contrasting effects of two identified amygdaloid efferent pathways on single hypothalamic neurons. J Neurophysiol 1986;31:37–248. 5. Canteras NS, Simerly RB, Swanson LW. Organization of projections from the medial nucleus of the amygdala: a PHAL study in the rat. J Comp Neurol 1995;360:213–245. 6. Herzog AG, Russell V, Vaitukaitis J. Neuroendocrine dysfunction in temporal lobe epilepsy. Arch Neurol 1982;39:133. 7. McEwen BS. How do sex and stress hormones affect nerve cells? Ann NY Acad Sci 1994;743:1–16. 8. Matsumoto A, (ed.) Sexual differentiation of the brain. Boca Raton, Fla: CRC Press, 1999. 9. Breedlove SM. Sexual dimporphism in the vertebrate nervous system. J Neurosci 1992;12:4133–4142. 10. Shughrue PJ, Lane MV, Merchenthaler I. Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central nervous system. J Comp Neurol 1997; 388:507–525. 11. Roselli CE, Resko JA. Cytochrome P450 aromatase (CYP19) in the non-human primate brain: distribution, regulation, and functional significance. J Steroid Biochem Mol Biol 2001;79:247–253. 12. LeVay SA. A difference in hypothalamic structure between heterosexual and homosexual me. Science 1991;253:1034–1037. 13. Zhou JN, Hofman MA, Gooren LJG, et al. A sex dif- ference in the human brain and its relation to transsex- uality. Nature 1995;378:68–70. 14. Harasty J, Double KL, Holliday GM, et al. Language- associated cortical regions are proportionally larger in the female brain. Arch Neurol 1997;54:171–176. 15. Rabinowicz T, MavDonald-Comber Petetot J, Gartside PS, et al. Structure of the cerebral cortex in men and women. J Neuropathol and Exp Neurol 2002;61:46– 57. 16. Angold A, Worthman CW. Puberty onset of gender dif- ferences in rates of depression: a developmental, epi- demiologic and neuroendocrine perspective. J Affect Dis- ord 1993;29:145–158. 17. Simerly RB, Chang M, Muramatsu, et al. Distribution of androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ hybridization study. J Comp Neuro 1990;294:76–95. 18. Wallis GJ, Luttge W. Influence of estrogen and proges- terone on glutamic acid decarboxylase activity in discrete regions of rat brain. J Neurochem 1980;34:609–613. 19. Murphy DD, Cole NB, Greenberger V, Segal M. Estra- diol increases dendritic spine density by reducing GABA neurotransmission in hippocampal neurons. J Neurosci 1998;18:2550–2559. 20. Pecins-Thompson M, Brown NA, Kohama SG, et al. Ovarian steroid regulation of tryptophan hydroxylase mRNA expression in rhesus macaques. J Neurosc 1996;16:7021–7029. 21. Briggs M. The relationship between monoamine oxidase activity and sex hormone concentrations in human blood plasma. J Reprod Fertil 1972;29:447–450. 22. Klein P, Versi E, Herzog AG. Mood and the menopause. Br J Obstet Gynaecol 1999;106:1–4. 23. Gibbs RB. Fluctuations of relative levels of choline acetyl transferase mRNA in different regions of the rat basal forebrain across the estrous cycle: effects of estrogen and progesterone. J Neurosci 1996;16:1049. 24. Luine VN. Estradiol increases choline acetyltransferase activity in specific basal forebrain nuclei and projection areas of female rat. Exp Neurol 1985;89:484. 25. Gibbs RB. Effects of estrogen on potassium-stimulated acetylcholine release in the hippocampus and overlying cortex of adult rats. Brain Res 1997;749:143–146. 26. Sherwin BB. Estrogen and cognitive aging in women. Trends Pharmacol Sci 2002;23:527–534. 27. Teresawa E, Timiras P. Electrical activity during the estrous cycle of the rat: cyclic changes in limbic struc- tures. Endocrinology 1968;83:207. 28. Klein P, Herzog AG. Hormonal effects on epilepsy in women. Epilepsia 1998;39(suppl 8):S9–S16. 29. Edwards HE, Burnham WM, Mendonca A, et al. Steroid hormones affect limbic afterdischarge thresholds and kindling rates in adult female rats. Brain Res 1999;838: 136–150. 30. Woolley DE, Timiras PS. The gonad-brain relationship: effects of female sex hormones on electroshock convul- sions in the rat. Endocrinol 1962;70:196–209. 31. Wong M, Moss R. Long-term and short-term electro- physiological effects of estrogen on the synaptic proper- ties of hippocampal CA1 neurons. J Neurosci 1992;12: 3217–3225. 32. Foy MR, Xu J, Xie X, et al. 17beta-estradiol enhances NMDA receptor-mediated EPSPs and long-term poten- tiation. J Neurophysiol 1999;81:925–929. 33. Woolley CS, McEwen BS. Role of estradiol and proges- terone in regulation of hippocampal dendritic spine den- sity during the estrous cycle in the rat. J Comp Neurol 1993;336:293–306. 34. Yankova M, Hart SA, Woolley CS. Estrogen increases synaptic connectivity between single presynaptic inputs and multiple postsynaptic CA1 pyramidal cells: a serial electron-microscopic study. Proc Natl Acad Sci USA 2001;98:3525–3530. 35. Landgren S, Aasly J, Backstrom T, et al. The effect of progesterone and its metabolites on the interictal epilep- tiform discharge in the cat’s cerebral cortex. Acta Phys- iol Scand 1987;131:33–42. 36. Backstrom T, Zetterlund B, Blom S, et al. Effects of intra- venous progesterone infusions on the epileptic discharge frequency in women with partial epilepsy. Acta Neurol Scand 1984;69:240–248. 37. Paul SM, Purdy RH. Neuroactive steroids. FASEB 1992;6:2311–2322. EFFECT OF OVARIAN HORMONES ON THE NERVOUS SYSTEM 89 38. Kokate TG, Banks, MK, Magee T, et al. Finasteride, a 5a-reductase inhibitor, blocks the anticonvulsant activ- ity of progesterone in mice. J Pharmacol Exp Ther 1999;288:679–684. 39. Majewska MD, Harrison NL, Schwartz RD, et al. Steroid hormone metabolites are barbiturate-like modulators of the GABA receptor. Science 1986;232:1004–1007. 40. Cheney DL, Uzunov D, Vosta E, et al. Gas chromato- graphic-mass fragmentographic quantitation of 3a- hydroxy-5a-pragnan-20-0ne (allopregnanolone) and its precursors in blood and brain of adrenalectomized and castrated rats. J Neurosci 1995;15:4641–4650. 41. Reddy DS, Kim HY, Rogawski MA. Neurosteroids with- drawal model of perimenstrual catamenial epilepsy. Epilepsia 2001;42:328–337. 42. Reddy DS, Rogawski MA. Enhanced anticonvulsant activity of neuroactive steroids in a rat model of cata- menial epilepsy. Epilepsia 2001;42:337–344. 43. Smith SS, Gong OH, Hsu FC, et al. GABA(A) receptor alpha-4 subunit suppression prevents withdrawal prop- erties of an endogenous steroid. Nature 1998;30:392: 926–930. 44. Reddy DS, Kulkami SK. Proconvulsant effects of neu- rosteroids pregnenolone sulfate and dehydroepiandros- terone sulfate in mice. Eur J Pharmacol 1998;345:55–59. 45. Kokate TG, Juhng KN, Krkby RD, et al. Convulsant actions of the neurosteroid pregnenolone sulfate in mice. Br Res 1999;831:119–124. 46. Flood JF, Morle JE, Roberts E. Pregnenolone sulfate enhances posttraining memory processes when injected in very low doses into limbic system structures. Proc Natl Acad Sci USA 1995;92:10806. 47. Morales AJ, Nolan J, Nelson G, et al. Effects of replace- ment dose of dehydroepiandrosterone (DHEA) in men and women of advancing age. J Clin Endocrinol Metab 1994;78:1360. 48. Wolfowitz OM, Reus VI, Roberts E, et al. Dehyepiandros- terone (DHEA) treatment of depression. Biol Psychiatry 1997;41:311. 49. Green PS, Simpkins JW. Neuroprotective effects of estro- gens: potential mechanisms of action. Int J Dev Neurosci 2000;18:347–358. 50. Garcia-Seguera LLM, Azcoitia I, Don Carlos LL. Neu- roprotection by estradiol. Prog Neurobiol 2001;63: 29–60. 51. Toran-Allerand CD, Guan X, MacLusky NJ, et al. ER-X: a novel, plasma membrane-associated, putative estrogen receptor that is regulated during development and after ischemic brain injury. J Neurosci 2002;22:8391–8401. 52. Gibbs RB. Estrogen replacement enhances acquisition of a spatial memory task and reduces deficits associated with hippocampal muscarinic receptor inhibition. Horm Behav 1999;36:222–233. 53. Horvath KM, Hartig W, Van der Veen R, et al. 17beta- estradiol enhances cortical cholinergic innervation and preserves synaptic density following excitotoxic lesions to the rat nucleus basalis magnocellularis. Neuroscience 2002;110:489–504. 54. Gibbs RB. Levels of trkA and BDNF mRNA, but not NGF mRNA, fluctuate across the estrous cycle and increase in response to acute hormone replacement. Brain Res 1998;810:294. 55. Gibbs RB. Treatment with estrogen and progesterone affects relative levels of brain-derived neurotrophic fac- tor mRNA and protein in different regions of the adult rat brain. Brain Res 1999;844:20–27. 56. Paganini-Hill A, Henderson VW. Estrogen replacement therapy and risk of Alzheimer disease. Arch Intern Med 1996;156:2213–2217. 57. Lerner AJ. Commentary:women and Alzheimer’s disease. J Clin Endocrinol Metab 1999;84:1830. 58. Wise PM, Wilson M, Dubal DB, Rau SW. In Vitro and in vivo approaches to the study of the neuroprotective actions of estradiol. In: Handa RJ, Hayashi S, Terasawa E, Kawata M, (eds.) Neuroplasticity, development and steroid hormone action. Boca Raton, Fla: CRC Press 2002;81–91. 59. Wise PM, Dubal DB, Wilson ME, et al. Estradiol is a pro- tective factor in the adult and aging brain: understand- ing of mechanisms derived from in vivo and in vitro stud- ies. Brain Res Brain Res Rev 2001;37:313–319. 60. Veliskova J, Velisek L, Galanopoulou AS, Sperber EF. Neuroprotective effects of estrogens on hippocampal cells in adult female rats after status epilepticus. Epilep- sia 2000;41(suppl 6):S30–35. 61. Beaulieu E, Schumacher M. Progesterone as a neuroac- tive neurosteroid, with special reference to the effect of progesterone on myelination. Steroids 2000;65:605–612. 62. Jay JR, MacLaughlin DT, Badger TM, et al. Hormonal modulation of Schwann cell tumors. I. The effects of estradiol and tamoxifen on methylnitrosourea-induced rat Schwann cell tumors. Ann NY Acad Sci 1986;486: 371–382. 63. Koenig HL, Ferzaz B, et al. Progesterone synthesis and myelin formation by Schwann cells. Science 1995;268: 1500–1503. 64. Murphy DG, DeCarli C, Daly E, et al. X-chromosome effects on female brain: a magnetic resonance imaging study of Turner’s syndrome. Lancet 1993;342: 1197–1200. 65. Hines M. Gonadal hormones and sexual differentiation of human behavior: effects on psychosexual and cogni- tive development. In: Matsumoto A, (ed.) Sexual Dif- ferentiation of the Brain . Boca Raton, Fla: CRC Press, 1999;257–278. 66. Klein P, Van Passell-Clark LM, Pezzullo JC. Onset of epilepsy at the time of menarche. Neurology 2003;60: 495–497. 67. Morrell MJ, Hamdy SF, Seale CG, Springer A. Self- reported reproductive history in women with epilepsy: puberty onset and effects of menarche and menstrual cycle on seizures. Neurology 1998;50(suppl 4):A448. 68. Rosciszewska D, The course of epilepsy in girls at the age of puberty. Neurol Neurochir Pol 1975;9:597–602. 69. Speroff L, Glass RH, Kase NG, (ed.) Clinical gynecologic endocrinology and infertility , 5th ed. Baltimore, Md: Willliams & Wilkins, 1994. 70. Genazzani AR, Bernardi F, Monteleone P, et al. Neu- ropeptides, neurotransmitters, neurosteroids, and the onset of puberty. Ann NY Acad Sci 2000;900:1–9. 71. Laidlaw J. Catamenial epilepsy. Lancet 1956;217: 1235–1237. 72. Backstrom T. Epileptic seizures in women related to plasma estrogen and progesterone during the menstrual cycle. Acta Neurol Scand 1976;54:321–347. 73. Mattson RH, Cramer JA, Caldwell BV. Seizure frequency and the menstrual cycle: a clinical study. Epilepsia 1981;22:242. 74. Herzog AG, Klein P, Ransill BJ.Three patterns of cata- menial epilepsy. Epilepsia 1997;38:1082–1088. 75. Burger HC. The endocrinology of the menopause. Matu- ritas 1996;23:129–136. NEUROLOGIC DISEASE IN WOMEN 90 76. Harden CL, Pulver MC, Ravdin L, Jacobs AR. The effect of menopause and perimenopause on the course of epilepsy. Epilepsia 1999;40:1402–1407. 77. Abbasi F, Krumholz A, Kittner SJ, Langenberg P. Effects of menopause on seizures in women with epilepsy. Epilepsia 1999;40:205–210. 78. Herzog A, Seibel M, Schomer DL, Vaitukaitis JL, Geschwind N. Reproductive endocrine disorders in women with partial seizures of temporal lobe origin. Arch Neurol 1986;43:341–346. 79. Klein P, Serje A, Pezzullo JC. Premature ovarian failure in women with epilepsy. Epilepsia 2001;42:1584–1589. 80. Drislane FW, Coleman AE, Schomer DL, et al. Altered pulsatile secretion of luteinizing hormone in women with epilepsy. Neurology 1994;44:306–310. 81. Stewart WF, Lipton RB, Celentano DD, et al. Prevalence of migraine headache in the United States. Relation to age, income, race, and other sociodemographic factors. JAMA 1992;267:64–69. 82. Ratinahirana H, Darbois Y, Bousser MG. Migraine and pregnancy: a prospective study in 703 women after deliv- ery. Neurology 1990;40(suppl 1):437. 83. Silberstein SD. Hormone-related headache. Med Clin North Am 2001;85:1017–1035. 84. Geary GG, Krause DN, Duckles SP. Estrogen reduces mouse cerebral artery tone through endothelial NOS- and cyclooxygenase-dependent mechanisms. Am J Phys- iol Heart Circ Physiol 2000;279:H511–519. 85. Krejza J, Mariak Z, et al. ICA flow correlates with serum estradiol levels during the menstrual cycle. Stroke 2001;32:30–36. 86. Epstein MT, Hockaday JM, Hockaday TD. Migraine and reproductive hormones throughout the menstrual cycle. Lancet 1975;1:543–548. 87. Limmroth V, Lee WS, Koskowitz MA. GABAA-recep- tor-mediated effects of progesterone, its ring-A-reduced metabolites and synthetic neuroactive steroids on neu- rogenic oedema in the rat meninges. Br J Pharmacol 1996;117:99–104. 88. Eckersell CB, Popper P, Micevych PE. Estrogen-induced alteration of mu-opioid receptor immunoreactivity in the medial [preoptic nucleus and medial amygdala. J Neu- rosci 1998;q8:3967–3976. 89. Cheung S, Salinas J, Hammer RP Jr. Gonadal steroid hor- mone-dependence of beta-endorphin-like immunoreac- tivity in the medial preoptic area of the rat. Brain Res 1995;675:83–88. 90. Dawson-Basoa M, Gintzer AR. Gestational and ovar- ian sex steroid antinociception: synergy between spinal kappa and delta opioid systems. Brain Res 1998;794: 61–67. 91. Herzog AG. Continuous bromocriptine therapy in men- strual migraine. Neurology 1997;48:101–102. 92. Zorgdrager A, DeKeyser J. Menstrually related worsen- ing of symptoms in multiple sclerosis. J Neurol Sci 1997;149:95–97. 93. McFarland HR. The management of multiple sclerosis. 3. Apparent suppression of symptoms by an estrogen- progestin compound. Mo Med 1969;66:209–211. 94. Confavreux C, Hutchinsoon M, Hours MM, et al. Rate of pregnancy-related relapse in multiple sclerosis. N Engl J Med 1998;339:285–291. 95. Riskind PN, Massacesi L, Doolittle TH, Hauser SL. The role of prolactin in autoimmune demyelination: sup- pression of experimental allergic encephalomyelitis by bromocriptine. Ann Neurol 1991;29:542–547. 96. Gilmore W, Weiner LP, Correale J. Effects of estradiol on cytokine secretion by proteolipid protein-specific T cell clones isolated from multiple sclerosis patients and nor- mal control subjects. J Immunol 1997;158:446–451. 97. Piccinni MP, Giudizi MG, Biagiotti R, et al. Progesterone favors the development of human T helper cells pro- ducing Th2-type cytokines and promotes both IL-4 pro- duction and membrane CD 30 expression in established Th1 cell clones. J Immunol 1995;155:128–133. 98. Pozzilli C, Falaschi P, Mainero C, et al. MRI in multiple sclerosis during the menstrual cycle: relationship with sex hormone patterns. Neurology 1999;53:622–624. 99. Bissay V, De Klippel N, Herroelen L, et al. Bromocrip- tine therapy in multiple sclerosis: an open label pilot study. Clin Neuropharmacol 1994;17:473–476. 100. Fleming JO, Hummel AL, Beinlich BR, et al. Vitamin D treatment of relapsing-remitting multiple sclerosis: a MRI-based pilot study. Neurology 2000;50(suppl 3)A:338. 101. Sicotte NL, Liva SM, Klutch R, et al. Treatment of mul- tiple sclerosis with the pregnancy hormone estriol. Ann Neurol 2002;52:421–428. 102. Bernardi M, Vergoni AV, Sandrini M, et al. Influence of ovariectomy, estradiol and progesterone on the behavior of mice in an experminetal model of depression. Phys- iol Behav 1989;45:1067–1068. 103. Girdler SS, Straneva PA, Light KC, Pedersen CA, Mor- row AL. Allopregnanolone levels and reactivity to men- tal stress in premenstrual dysphoric disorder. Biol Psy- chiatry 2001;49:788–797. 104. Nappi RE, Petraglia F, Luisi S, Polatti F, Farina C, Genaz- zani AR. Serum allopregnanolone in women with post- partum “blues.” Obstet Gynecol 2001;97:77–80. 105. Thulin PC, Carter JH, Nichols MD, et al. Menstrual- cycle related changed in Parkinson’s Disease. Neurology 1996;46(suppl)A:376. 106. Omdal R, Roalso S. Chorea gravidarum and chorea associated with oral contraceptives—diseases due to antiphospholipid antibodies? Acta Neurol Scand 1992; 86:219–220. 107. Baulieu EE. Neurosteroids: of the nervous system, by the nervous system, for the nervous system. Recent Prog Horm Res 1997;52:1–32. or the most part, genetic diseases do not discriminate between the sexes, affecting both men and women with equal severity and in a similar man- ner. In a small number of heritable disorders, however, special considerations arise in the clinical management of women that differ considerably from those in men. Uniquely, in genetics, the clinician is concerned about manifestations not only in the patient but also in her relatives, especially actual or potential offspring to whom disease may be transmitted. Some genetic diseases are different in women than in men. In others, the only difference is in transmission; the offspring of an affected woman being at different risk than those of an affected man. In still others, notably the sex-linked disorders, both the disease and transmission pattern are different in men and women. EXPRESSION OF GENETIC DISEASES IN WOMEN Gender differences in disease phenotype are either sex- linked, the underlying gene(s) being located on a sex chro- mosome, or sex-limited autosomal disorders, such as male-pattern baldness. In theory, sex-linked disorders could result from alterations of either the X or the Y chro- mosome. However, the Y chromosome is not only small but also has a low density of genes (1). Its known con- tribution to human neurogenetic disorders appears to be limited to a behavioral and mildly dysmorphic phenotype, the XYY syndrome (2). In practice, virtually all sex-linked disorders are encoded by genes on the X chromosome. Sex-Linked Disorders Recognized X-linked disorders are slightly more frequent than would be predicted by the ratio of one X chromo- some to 22 autosomes. As of this writing, of the 14,561 entries in the catalog of human genes and genetic disor- ders, Online Mendelian Inheritance in Man (OMIM), 810 (5.56% of the total) are in the X chromosome catalog (1). Indeed, 101 of the 1,348 phenotype descriptions in OMIM are in the X chromosome catalog, representing 7.49% of the total number of all phenotypic descriptions in the entire catalog. This is a higher percentage than would be expected from the relative size of the X chro- mosome, the 151,567,156 base pairs (bp) of which rep- resent only 4.67% of the 3,242,415,757 bp that consti- tute the haploid human genome. Indeed, Ensembl, a joint project between the European Bioinformatics Institute and the Sanger Institute, currently predicts the existence of 24,847 human genes, of which 869 (only 3.49%) are on the X chromosome (3). Thus, there are roughly twice as many known human X-linked traits as would be pre- dicted by the proportion of human genes that is currently 91 Genetic Disorders in Women Orest Hurko, MD 7 F [...]... syndrome 30 0100 30 335 0 31 2920 30 8750 Xq28 Xq28 Xq22 X? Peroxisomal ATP-binding transport protein L-CAM cell adhesion molecule Proteolipid protein PRODUCT PROGRESSIVE ATAXIAS DISORDER OMIM LOCUS GENE Menkes kinky-hair disease 30 9400 Pelizaeus-Merzbacher PRPS deficiency X-linked cerebellar ataxia (X-linked OPCA included) X-linked ataxia-deafness syndrome Arts fatal X-linked ataxia syndrome 31 2080 31 1850 30 2500... 31 0200 31 030 0 Xp21.1 Xq28 Dystrophin Emerine, serine-rich vesicular transport protein 30 9660 X 31 0400 Xq28 30 5550 31 1800 X 31 1800 Fingerprint myopathy Phosphoglycerate kinase deficiency PRODUCT MTM1 myotubularin, putative tyrosine phosphatase PGK-I PERIPHERAL NEUROPATHIES DISORDER OMIM LOCUS GENE Charcot-Marie-Tooth, X-linked dominant (CMTX1) Charcot-Marie-Tooth, X-linked recessive (CMTX2) Charcot-Marie-Tooth,... Charcot-Marie-Tooth, X-linked recessive (CMTX3) Charcot-Marie-Tooth, 2D (Cowchock variant with deafness and mental retardation) Charcot-Marie-Tooth, with deafness and optic atrophy (Rosenberg-Chutorian disease) Charcot-Marie-Tooth, with aplasia cutis congenita Fabry disease (angiokeratoma diffusa) 30 2800 XP11 .3 Connexin 32 , gap junction protein 30 2801 Xp22.2 30 2802 Xp26 31 0490 Xq24-q26 1 31 1070 X 30 28 03 301500... H Sex-linked recessive inheritance in Charcot-Marie-Tooth disease with partial manifestation in female carriers Hum Genet 1980;55: 4 13 415 85 Hanemann CO, Bergmann C, Senderek J, et al Transient, recurrent, white matter lesions in X-linked Charcot-Marie-Tooth disease with novel connexin 32 mutation Arch Neurol 20 03; 60:605–609 86 Bergoffen J, Scherer SS, Wang S, et al Connexin mutations in X-linked... no report of clinical or subclinical neurologic involvement in true female carriers in this disorder or in the other X-linked motor neuron disease, lethal infantile sex-linked spinal muscular atrophy (SMAX2) (1 03) .] Motor neuron disease may underly some forms of distal infantile arthrogryposis, of which there may be as many as three distinct X-linked types (104) In one such family, the disease was transmitted... ganglia disease, Dandy-Walker malformation with mental retardation and seizures HHHH syndrome (hereditary hemihypotrophy hemiparesis hemiathetosis) Goeminne TKCR syndrome (torticollis, keloids, cryptorchidism, renal dysplasia) 31 4250 Xq12-q 13. 1 31 1510 Xq28 30 434 0 Xq25-q27 30 6960 X? 31 430 0 Xq28 PRODUCT (continued) NEUROLOGIC DISEASE IN WOMEN 108 TABLE 7.2 X-Linked Neurogenetic Disorders Seen in Males... 31 2080 31 1850 30 2500 Xq 13. 2q 13. 3 Xq28 Xq22-q24 X Cu(2+)-transporting ATPase, alpha polypeptide Proteolipid protein Phosphoribosyl pyrophosphate synthetase 30 1790 30 1 835 X X PRODUCT MOVEMENT DISORDERS DISORDER OMIM LOCUS GENE Lesch-Nyhan syndrome 30 8000 X Hypoxanthine-guanine phosphoribosyltransferase Torsion-dystonia 3 with parkinsonism, Filipino type Waisman early-onset parkinsonism and mental retardation... 170 Machin GA, Walther GL, Fraser VM, et al Autopsy findings in two adult siblings with Coffin-Lowry syndrome Am J Med Genet 1987;(suppl 3) :30 3 30 9 171 Simensen R J, Abidi F, Collins JS, et al Cognitive function in Coffin-Lowry syndrome Clin Genet 2002;61: 299 30 4 172 Sivagamasundari U, Fernando H, Jardine P, et al The association between Coffin-Lowry syndrome and psychosis: a family study J Intellect... X-linked Charcot-Marie-Tooth disease Science 19 93; 262:2 039 –2042 87 Ionasescu VV, Trofatter J, Haines JL, et al X-linked recessive Charcot-Marie-Tooth neuropathy: clinical and genetic study Muscle Nerve 1992;15 :36 8 37 3 88 Cowchock FS, Duckett SW, Streletz LJ, et al X-linked motor-sensory neuropathy type-II with deafness and mental retardation: a new disorder Am J Med Genet 1985;20 :30 7 31 5 89 Priest JM,... Hyperuricemia and neurological deficits: a family study N Engl J Med 1970;282:992–997 136 Schmidley JW, Levinsohn MW, Manetto CV, et al Infantile X-linked ataxia and deafness: a new clinicopathologic entity Neurology 1987 ;37 : 134 4– 134 9 137 Arts WFM, Loonen MCB, Sengers RCA, et al X-linked ataxia, weakness, deafness, and loss of vision in early childhood with a fatal course Ann Neurol 19 93; 33: 535 – 539 138 Shokeir . medi- ated by its 3 ␣-hydroxylated metabolite, 3- -hydroxy- 5- - pregnan-20-one or allopregnanolone (AP) (37 ,38 ). Allo- pregnanolone and the 3 ,5␣-hydroxylated natural metabolite of the mineralocorticoid. Estradiol is a pro- tective factor in the adult and aging brain: understand- ing of mechanisms derived from in vivo and in vitro stud- ies. Brain Res Brain Res Rev 2001 ;37 :31 3 31 9. 60. Veliskova. report of clinical or subclinical neuro- logic involvement in true female carriers in this disorder or in the other X-linked motor neuron disease, lethal infan- tile sex-linked spinal muscular

Ngày đăng: 10/08/2014, 00:20

Tài liệu cùng người dùng

Tài liệu liên quan