Neurourology

Published on 12/04/2015 by admin

Filed under Neurology

Last modified 12/04/2015

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 2828 times

Chapter 38 Neurourology

Urogenital dysfunction can result from a wide range of neurological conditions, and the importance of this problem to the patient’s health and its negative impact on quality of life and loss of dignity is now widely recognized. The investigations and management of disorders of urogenital function was formerly regarded as the preserve of urologists. But as neurologists and rehabilitation specialists become aware of the range of possible effective nonsurgical treatments and increasingly inquire after patients’ complaints of disordered urogenital function, they are taking a more active interest in uroneurology—bladder dysfunction viewed from a neurological perspective. This chapter describes what a neurologist needs to know for the management of patients with neurogenic urogenital problems. Urodynamic, neurophysiological, and radiological investigations and available medical treatments are described.

Lower Urinary Tract and Its Neurological Control

The lower urinary tract consists of the bladder and urethra and has two roles: storage of urine and voiding at appropriate times. Control of the detrusor and urethral sphincter muscles in these two mutually exclusive states is dependent upon both local spinal reflexes and central cerebral control. The pontine micturition center, which receives input from higher centers (including the periaqueductal gray of the midbrain, hypothalamus, and cortical areas such as the medial prefrontal cortex) is responsible for switching between the two states. The frequency of micturition in a person with a bladder capacity of 400 to 600 mL is once every 3 to 4 hours. Voiding takes 2 to 3 minutes, so this means that for more than 98% of life, the bladder is in a storage phase. Switching to a voiding phase is initiated by a conscious decision triggered by the perceived state of bladder fullness and an assessment of the social appropriateness of doing so. To effect both storage and voiding, connections between the pons and the sacral spinal cord must be intact, as must the peripheral innervation that originates from the most caudal segments of the cord. During the storage phase, sympathetic- and pudendal-mediated contraction of the internal and external urethral sphincters, respectively, maintains continence. Inhibition of the parasympathetic outflow prevents detrusor contractions (Fowler et al., 2008). When it is deemed appropriate to void, the pontine micturition center is no longer tonically inhibited, and reciprocal activation-inhibition of the sphincter-detrusor reverses. Relaxation of the pelvic floor and external and internal urethral sphincters, accompanied by parasympathetic-mediated detrusor contraction, results in effective bladder emptying. Intact neural circuitry between the pontine micturition center and bladder ensures coordinated activity between the detrusor and sphincter muscles. Fig. 38.1 reviews the innervation of the bladder.

image

Fig. 38.1 Innervation of lower urinary tract. A, Sympathetic fibers (shown in blue) originate in T11-L2 spinal cord segments and run through inferior mesenteric ganglia (inferior mesenteric plexus [IMP]) and hypogastric nerve (HGN) or through the paravertebral chain to enter pelvic nerves at base of bladder and urethra. Parasympathetic preganglionic fibers (shown in green) arise from S2-S4 spinal segments and travel in sacral roots and pelvic nerves (PEL) to ganglia in the pelvic plexus (PP) and bladder wall. This is where the postganglionic nerves that supply parasympathetic innervation to the bladder arise. Somatic motor nerves (shown in yellow) that supply striated muscles of external urethral sphincter arise from S2-S4 motor neurons and pass through pudendal nerves. B, Efferent pathways and neurotransmitter mechanisms that regulate lower urinary tract. Parasympathetic postganglionic axons in pelvic nerve release acetylcholine (ACh), which produces bladder contraction by stimulating M3 muscarinic receptors in bladder smooth muscle. Sympathetic postganglionic neurons release noradrenaline (NA), which activates β3-adrenergic receptors to relax bladder smooth muscle and activates β1-adrenergic receptors to contract urethral smooth muscle. Somatic axons in pudendal nerve also release Ach, which produces contraction of external sphincter striated muscle by activating nicotinic cholinergic receptors. L1, First lumbar root; S1, first sacral root; SHP, superior hypogastric plexus; SN, sciatic nerve; T9, ninth thoracic root.

(From Fowler, C.J., Griffiths, D., de Groat, W.C., 2008. The neural control of micturition. Nat Rev Neurosci 9, 453-466.)

Functional brain imaging studies have demonstrated that neurological control of the bladder in humans is essentially similar to that demonstrated in experimental animals. A number of positron emission tomography (PET) and, more recently, functional magnetic resonance imaging (fMRI) studies have investigated human control of urinary storage and voiding. The initial PET experiments of Blok and colleagues identified the brain centers activated during attempted micturition (Blok et al., 1997, 1998). In those able to void during the scanning, activity was shown in a region of the medioposterior pons called the M-region. In those subjects unable to void, a distinct region in the ventrolateral pontine tegmentum was activated, the L-region. Although it had been demonstrated in cats that separate pontine nuclei exist for the storage and voiding phases of bladder activity, subsequent experiments have failed to consistently demonstrate activity in this distinct L-region. In the cortex, the PET scans showed significant activity in the right inferior frontal gyrus and the right anterior cingulate gyrus during voiding that was not present during the withholding phase. Nour and associates then corroborated these findings with their own PET study of 12 healthy male volunteers, showing activity in a number of brain areas including the cerebellum (Nour et al., 2000). Other areas that show activation in fMRI during bladder filling include the anterior cingulate gyrus and right insula; fMRI has shown that the medial prefrontal cortex, responsible for complex cognitive and socially appropriate behavior, plays an important role in voiding. A recent article reviews the functional imaging studies evaluating bladder functions and their contributions to our understanding of bladder control (Fowler and Griffiths, 2010).

Bowel and Its Neurological Control

Similar to the bladder, the lower bowel also exists mostly in the storage mode. Continence is maintained by a combination of the acute anorectal angle, maintained by puborectalis contraction, and internal anal sphincter tone, determined by sympathetic activity. In health, defecation can be delayed if necessary by contraction of the external anal sphincter and pelvic floor musculature, which requires sensory feedback from the anorectum. The process of defecation involves a series of neurologically controlled actions that begin in response to the conscious sensation of a full rectum. When this is perceived and if judged to be appropriate, defecation is initiated by maneuvers to raise the intraabdominal pressure and by straining down, causing descent of the pelvic floor. The internal anal sphincter pressure falls as a result of the rectoanal inhibitory reflex, and the pubococcygeus and striated external sphincter muscles relax. Functional imaging has been applied to evaluate central processing of different types of gastrointestinal (GI) stimulation. Hobday and associates used fMRI to identify the brain centers involved in the processing of anal (somatic) and rectal (visceral) sensation in healthy adults (Hobday et al., 2001). Rectal stimulation produced activation of somatosensory cortex, insula, anterior cingulate, and prefrontal cortex (PFC); anal canal stimulation produced similar regions of activity, although anterior cingulate activity was absent, and the primary somatosensory activation was slightly more superior in location. The activation of cingulate cortex with rectal stimulation may signify the function of the limbic system in the processing of visceral stimuli.

The processing of rectal sensation is relevant in bladder function because unlike other gut organs, it has an important sensory role, and the rectum is a visceral organ that contains both unmyelinated C fibers and thinly myelinated Aδ afferents. In contrast, the anal canal has somatic innervation from the pudendal nerve. The study by Hobday and associates has highlighted the differences in its cortical representation from that of the rectum. The various brain imaging studies of visceral stimulation, including the foregoing report, have been reviewed (Derbyshire, 2003). When different visceral stimuli—esophageal distension, esophageal pain, rectal distension, or the mechanisms operative in irritable bowel syndrome—were analyzed, esophageal stimulation was found to activate the insula most consistently, with other commonly involved areas including somatosensory and motor cortices. Considerable variation was observed in whether the periaqueductal gray was activated or not. Lower GI stimuli predominantly activated the prefrontal and orbitofrontal cortices as well as the insula, with variability in cingulate activation. Overall, esophageal stimulation involved a more central sensory and motor neural circuit, whereas lower GI stimulation activated areas with projections to autonomic and affective control centers such as the brainstem and amygdala.

Sexual Function and Its Neurological Control

Physiological sexual response in men and women has been divided classically into four phases: excitement, plateau, orgasm, and resolution. Excitation occurs in response to either physical or psychological stimulation and results in penile or clitoral tumescence and erection or vaginal lubrication. The plateau phase is accompanied by the various physical changes of high sexual arousal in anticipation of orgasm. Orgasm, an intensely sensory event, usually is associated with rhythmic contraction of the pelvic floor and culminates with ejaculation in men. During resolution, the increased genital blood flow resolves. A modification of this model of sexual response was the three-phase model proposed by Kaplan, consisting of desire, arousal, and orgasm.

Much remains to be discovered about cortical control of sexual function. Although it is thought that cerebral processing determines libido and desire, the ability to effect a sexual response is determined by spinal autonomic reflexes. Libido is hormone dependent with a major hypothalamic component, and loss of libido may be the earliest symptom of a pituitary tumor. In experimental animals, the deep anterior midline structures that form the limbic system have been shown to be important for sexual responses, and the medial preoptic–anterior hypothalamic area has an integrating function (Andersson, 2001). Functional brain imaging experiments including five PET studies and seven fMRI studies have highlighted the key areas of brain activity associated with sexual functioning—for example, the role of the hypothalamus in reproductive function, regulation of human sexuality, and regulation of erection through medial preoptic area, or the roles of the insula and claustrum in autonomic regulation and visceral sensory processing. The aspects of sexual function covered include penile sexual stimulation (Arnow et al., 2002; Georgiadis and Holstege, 2005), male ejaculation (Holstege et al., 2003), and visual sexual stimuli (Karama et al., 2002).

Male Sexual Response

Erection results from increased blood flow into the corpora cavernosa caused by relaxation of the smooth muscle in the cavernosal arteries and a reduction in venous return. The major peripheral innervation determining this is parasympathetic, which arises from the S2-S4 segments and travels to the genital region in the pelvic nerves. Sympathetic input is also important: sympathetic innervation of the genital region originates in the thoracolumbar chain (T11-L2) and travels through the hypogastric nerves to the confluence of nerves that lies on either side of the rectum and the lower urinary tract—the pelvic plexus.

The pelvic plexus also receives input from the pelvic nerves. It is from the pelvic plexus that the cavernous nerves arise and innervate the corpora cavernosa. Although erection is induced by parasympathetic activity, nitric oxide has been identified as important in causing relaxation of the corporeal blood vessels and the increase in penile blood flow that causes erection. Psychogenic erection requires cortical activation of spinal pathways, and the preservation of this type of responsiveness in men with low spinal cord lesions suggests that sympathetic pathways can mediate it. Reflex erections occur as the result of cutaneous genital stimulation. Preservation of reflex erections in men with lesions above T11 indicates that the response is the result of spinal reflexes, with afferent signals conveyed in the pudendal nerve and S2-S4 roots, and efferent signals through the same sacral roots. In health, reflex and psychogenic responses are thought to reinforce one another. In men, orgasm and ejaculation are not the same process; ejaculation is the release of semen, and orgasm consists of the sensory changes accompanied by pelvic floor contractions. Ejaculation involves emission of semen from the vas and seminal vesicles into the posterior urethra and closure of the bladder neck. The latter processes are under sympathetic control, whereas contraction of the pelvic floor muscles is under somatic nerve control, innervation being from the perineal branch of the pudendal nerve. After ejaculation, a period of resolution is necessary before sexual activity can be reinitiated.

Neurogenic Bladder Dysfunction

Lesions of the nervous system, central or peripheral, can result in characteristic patterns of bladder dysfunction depending upon the level of the lesions in the neurological axis (Panicker et al., 2010). The storage function of the bladder is affected following suprapontine or infrapontine/suprasacral lesions. This results in involuntary spontaneous or induced contractions of the detrusor muscle (detrusor overactivity), which can be identified during the filling phase of urodynamics. The voiding function of the bladder can be affected by infrapontine lesions. Following spinal cord damage, there is simultaneous contraction of the external urethral sphincter and detrusor muscle, detrusor-sphincter dyssynergia, which results in incomplete bladder emptying and abnormally high bladder pressures. Following lesions of the conus medullaris or cauda equina, voiding dysfunction can be due to poorly sustained detrusor contractions and possibly non-relaxing urethral sphincters (Table 38.1).

Cortical Lesions

Bladder Dysfunction

It has been known since the 1960s that anterior regions of the frontal lobes are critical for bladder control. Among patients with disturbed bladder control, various frontal lobe disturbances have been reported: intracranial tumors, damage after rupture of an aneurysm, penetrating brain wounds, and prefrontal lobotomy (leukotomy). The typical clinical picture of frontal lobe incontinence is a patient with severe urgency and frequency of micturition and urge incontinence but without dementia; the patient is socially aware and embarrassed by the incontinence. Micturition is normally coordinated, indicating that the disturbance is in the higher control of these processes. Urinary retention also has been described in patients with brain lesions. A small number of case histories have described patients with right frontal lobe disorders who had urinary retention and in whom voiding was restored when the frontal lobe disorder was treated successfully (Fowler, 1999).

Urinary incontinence develops in some patients after stroke. Urodynamic studies in incontinent patients have been carried out, and the general conclusion drawn from studying patients with disparate cortical lesions is that voiding mostly is normally coordinated. The most common cystometric finding is that of detrusor overactivity. It has not been possible to demonstrate a correlation between any particular lesion site and urodynamic findings. Urinary incontinence at 7 days following stroke predicts poor survival, disability, and institutionalization independent of level of consciousness. It has been suggested that incontinence in such cases is the result of severe general loss of function, or that persons who became incontinent may be less motivated to recover both continence and general function. Patients with hemorrhagic stroke are more likely to have detrusor underactivity in urodynamics compared to patients with ischemic stroke, who more often have detrusor overactivity (Han et al., 2010). Small-vessel disease of the white matter (leukoaraiosis) is associated with urgency incontinence, and it is increasingly becoming apparent that this is an important cause for incontinence in the functionally independent elderly (Tadic et al., 2010).

The cause of urinary incontinence in dementia is probably multifactorial. Not all incontinent older adults are cognitively impaired, and not all cognitively impaired older adults are incontinent. In a study of patients with progressive cognitive decline, incontinence was observed to occur in more advanced stages of Alzheimer disease (AD), whereas it could occur earlier on in the course of patients with dementia with Lewy bodies (DLB) (Ransmayr et al., 2008).

A much less common cause of dementia is normal-pressure hydrocephalus, where incontinence is a cardinal feature. Improvement in urodynamic function has been demonstrated within hours of lumbar puncture (LP) in patients with this disorder.

Sexual Dysfunction

Before functional imaging experiments, all that was known about human cerebral control and sexuality came from observations of patients with brain lesions, particularly those affecting temporal or frontal regions. These areas can be involved by disorders that cause epilepsy or by trauma, tumors, cerebrovascular disease, or encephalitis. It has long been observed that sexual dysfunction is more common in men and women with epilepsy. Although various sexual perversions and occasionally hypersexuality have been described in patients with temporal lobe epilepsy (TLE), the picture most commonly seen is that of sexual apathy. From studies comparing sexual dysfunction in generalized epilepsy with that in focal TLE, the evidence is sufficient to suggest that the deficit is a result of the specific temporal lobe involvement rather than a consequence of epilepsy, psychosocial factors, or antiepileptic medication. The problem usually is that of a low or absent libido, of which patients may not complain.

The role of hormonal dysfunction has yet to be fully determined. On the basis of measurements of sex hormones and pituitary function, it has been suggested that the hyposexuality of TLE results from a subclinical hypogonadotropic hypogonadism, and that dysfunction of medial temporal lobe structures may dysmodulate hypothalamopituitary secretion (Murialdo et al., 1995). Erectile dysfunction (ED) with preservation of libido can occur in men with temporal lobe damage with or without epilepsy and may be characterized by loss of nocturnal penile tumescence. Surgery for epilepsy rarely restores erectile function, although a survey of operated patients showed a higher level of satisfaction with sexual function among those who were free of seizures. Sexual dysfunction is common after head injury, particularly in patients who demonstrate cognitive damage. Hypersexual behavior may occur after frontal lobe damage. Lesions of the frontal lobes, the basal-medial part in particular, may lead to loss of social control, which also may affect sexual behavior.

Basal Ganglia Lesions

Bladder Dysfunction

Bladder symptoms in Parkinson disease (PD) correlate with neurological disability (Araki and Kuno, 2000) and stage of disease; both findings appear to support a link between dopaminergic degeneration and symptoms of urinary dysfunction. In line with current thinking about staging of PD in terms of underlying neuropathology (Braak et al., 2004), it appears that bladder dysfunction does not occur until some years after the onset of motor symptoms, and the dysfunction is correlated with the extent of dopamine depletion (Sakakibara et al., 2001b). This means that the underlying pathological process is likely to have extended into the neocortex and explains why the clinical context in which bladder dysfunction is seen in PD is common in patients with cerebral symptoms, as well as with adverse effects of long-standing treatment with dopaminergic agents.

The most frequent complaints are of nighttime frequency, urgency, and difficulty voiding (Sakakibara et al., 2001a), and the most common abnormality in urodynamic studies is detrusor overactivity (Araki et al., 2000). Of the several possible explanations for this finding, the hypothesis most widely accepted is that in healthy persons, the basal ganglia has an inhibitory effect on the micturition reflex, and with neuronal loss in the substantia nigra, detrusor overactivity develops. Studies on anesthetized cats demonstrated that rhythmic bladder contractions were inhibited by intracerebroventricular administration of a dopamine D1 receptor agonist but were not affected by a D2 receptor agonist. From this evidence, it was concluded that the D1 receptor provides the main inhibitory influence on the micturition reflex (Yoshimura et al., 2003). Clinical studies that have looked at the effect of l-dopa or apomorphine on bladder behavior in patients with PD have, however, produced conflicting results. In patients demonstrating the on/off phenomenon, cystometry done after taking l-dopa showed improvement of overactivity in some and worsening in others. The unpredictable effect of antiparkinsonian medications on urinary symptoms has not been definitively shown to correlate with age or stage of disease (Winge and Fowler, 2006). Many patients with PD have nocturnal polyuria as well, which contributes to bladder symptoms and would not be expected to improve with antimuscarinics. In addition to neurogenic bladder dysfunction, benign prostatic obstruction may occur concomitantly in some men with PD and contribute to bladder dysfunction. A recent study suggests that contrary to previous teaching, transurethral prostate resection may be successful in carefully selected PD men and is associated with minimal risk of incontinence (Roth et al., 2009).

In a patient with severe urinary symptoms but mild parkinsonism, a diagnosis of multiple system atrophy (MSA) should be considered. The onset of urogenital symptoms in MSA may precede overt neurological involvement; ED and bladder symptoms begin on average 4 to 5 years before diagnosis and 2 years before more specific neurological symptoms appear. The neuronal degeneration of MSA affects the central nervous system (CNS) at several locations that are important for bladder control, which probably explains why urinary complaints occur so early and are so severe in this condition. It is thought that detrusor overactivity is caused by neuronal loss in the pontine region, whereas incomplete bladder emptying is caused by loss of parasympathetic innervation of the detrusor after neuronal degeneration in the intermediolateral cell columns of the spinal cord. In addition, anterior horn cell loss in the Onuf nucleus results in denervation of the urethral sphincter so that the patient has a combination of detrusor overactivity, incomplete bladder emptying, and a weak sphincter. Bladder dysfunction may change during the progression of MSA, and serial studies have shown that the mean postmicturition residual volume increases as the condition progresses (Ito et al., 2006). Bladder symptoms in other parkinsonian syndromes are less prominent than in MSA, and although they may occur as part of the patient’s general disability, they rarely are as severe as in MSA and do not occur at a stage of the disease when a neurological cause is not evident.

Bowel Dysfunction

Constipation is now thought to be a preclinical manifestation of PD, and a symptom questionnaire showed that this was considered to be the most bothersome nonmotor symptom for PD patients (Sakakibara et al., 2001a). Several possible causes for constipation are recognized: a slow colonic transit time has been demonstrated in a number of studies; this finding may be secondary to a reduction in dopaminergic myenteric neurons. An abnormality of the defecation process has also been demonstrated in some patients with PD, with paradoxical contraction of the external anal sphincter and pubococcygeus causing outlet obstruction. This phenomenon can result in anismus and is thought to be a form of focal dystonia. Bowel dysfunction appears earlier and progresses faster in patients with MSA than in those with PD (Stocchi et al., 2000).

Sexual Dysfunction

Experimental evidence from animals and humans shows that dopaminergic mechanisms are involved in determining libido and inducing penile erection. In animal studies, the medial preoptic area of the hypothalamus has been shown to regulate sexual drive, and selective stimulation of dopamine D2 receptors in this region increases sexual activity in rats (Andersson, 2001). An increase in libido in some patients with PD treated with dopamine agonists and l-dopa as part of the “hedonistic homeostatic dysregulation” syndrome (Giovannoni et al., 2000) is a well-recognized phenomenon, although its incidence is uncertain. The cause of ED in PD is unclear, but it is a significant problem and in one study was shown to affect 60% of a group of men with PD, compared with 37.5% of age-matched healthy men. ED usually affects men later in the course of PD, with onset years after the diagnosis of neurological disease has been established. A survey of relatively young patients with PD (mean age, 49.6 years) and their partners revealed a high level of sexual dysfunction, with the most severely affected couples being those in which the patient was male.

ED may be the first symptom in men with MSA, predating the onset of any other neurological symptoms by several years. The disorder appears to be chronologically distinct from the development of postural hypotension. The reason for the apparently early selective involvement of neural mechanisms for erection is not known. Preserved erectile function in a man with parkinsonism strongly contradicts the diagnosis of MSA. The available literature on female sexual problems in movement disorders is limited (Jacobs et al., 2000; Oertel et al., 2003).

Brainstem Lesions

Voiding difficulty is a rare but recognized symptom of a posterior fossa tumor and has been reported in series of patients with brainstem disorders (Fowler, 1999). In an analysis of urinary symptoms of 39 patients who had had brainstem strokes, lesions that resulted in micturition disturbance usually were dorsally situated (Sakakibara et al., 1996)—a finding consistent with the known location of the brainstem centers involved in bladder control. The proximity in the dorsal pons between the pontine micturition center and medial longitudinal fasciculus means that a disorder of eye movements, such as an internuclear ophthalmoplegia (INO), is highly likely in patients with a pontine disorder causing a voiding difficulty.

Spinal Cord Lesions

Bladder Dysfunction

Spinal cord disorders are the most common cause for neurogenic bladder dysfunction. Transspinal pathways connect the pontine micturition centers to the sacral cord. Intact connections are necessary to effect the reciprocal activity of the detrusor and sphincter needed to switch between storage and voiding. After disconnection from the pons, this synergistic activity is lost, resulting in detrusor-sphincter dyssynergia.

Initially after acute SCI, there usually is a phase of neuronal shock of variable duration, characterized clinically by complete urinary retention and urodynamics demonstrating an acontractile detrusor. Gradually over the course of weeks, new reflexes emerge to drive bladder emptying and cause detrusor contractions in response to low filling volumes. The neurophysiology of this recovery has been studied in cats, and it has been proposed that after spinal injury, C fibers emerge as the major afferents, forming a spinal segmental reflex that results in automatic voiding. It is assumed that the same pathophysiology occurs in humans. In support of this assumption is the observed response to intravesical capsaicin (a C-fiber neurotoxin) in patients with acute traumatic spinal cord injury (SCI) or chronically progressive spinal cord disease from multiple sclerosis (MS). The abnormally overactive, small-capacity bladder that characterizes spinal cord disease causes patients to experience urgency and frequency. However, patients with complete transection of the cord may not complain of urinary urgency. If detrusor overactivity is severe, incontinence is highly likely. Poor neural drive on the detrusor muscle during attempts to void, together with an element of detrusor-sphincter dyssynergia, contributes to incomplete bladder emptying. This difficulty may exacerbate the symptoms of the overactive bladder. Although the neurological process of voiding may have been as severely disrupted as the process of storage, the symptoms of difficulty emptying can be minor compared with those of urge incontinence. Only on direct questioning may the patient admit to having difficulty initiating micturition, an interrupted stream, or possibly a sensation of incomplete emptying.

Because bladder innervation arises more caudally than innervation of the lower limbs, any form of spinal cord disease that causes bladder dysfunction is likely to produce clinical signs in the lower limbs as well, unless the lesion is limited to the conus. This rule is sufficiently reliable to be of great value in determining whether a patient has a neurogenic bladder caused by spinal cord disease.

Multiple Sclerosis

The pathophysiological consequences of progressive MS affecting the spinal cord for the bladder are similar to those of SCI, but the medical context of increasing disability is such that management must be quite different. Estimates of the proportion of patients with MS who have lower urinary tract symptoms vary according to the severity of the neurological disability in the group under study, but a figure of about 75% is frequently cited (Marrie et al., 2007). Several studies have shown that urinary incontinence is considered to be one of the worst aspects of the disease; 70% of a self-selected group of patients with MS responding to a questionnaire classified the impact bladder symptoms had on their life as “high” or “moderate” (Hemmett et al., 2004). A strong association between bladder symptoms and the presence of clinical spinal cord involvement, including paraparesis and UMN signs, has been recognized on examination of the lower limbs in patients with MS. This observation has also been made in patients with a similar condition, acute disseminated encephalomyelitis (ADEM) (Panicker et al., 2009).

The most common urinary symptom is urgency; all series of urodynamic studies in patients with MS have shown that this is due to detrusor overactivity. Hesitancy of micturition may be a symptom patients volunteer or admit on direct questioning, but the more disabled may find themselves unable to initiate micturition voluntarily, emptying their bladders only with an involuntary hyperreflexic contraction and an interrupted urinary flow. Evidence of incomplete emptying may come not from a sensation of continued fullness after voiding but rather from the need to pass urine again within 5 to 10 minutes (double voiding).

As the neurological condition progresses, bladder dysfunction may become more difficult to treat. However, unlike with the bladder dysfunction that follows SCI, progressive neurological diseases such as MS rarely result in upper urinary tract involvement. This is the case even when long-standing MS has resulted in severe disability and spasticity. The reason for this sparing of the upper urinary tract is not known, but it means that in such patients, management should emphasize symptomatic relief (Fowler, 1999).

A particular problem in MS is that neurological symptoms may deteriorate acutely when the patient has an infection and pyrexia, including urinary tract infection (UTI). As MS progresses, recurrent infections are likely to result in deficits that accumulate and lead to progressive neurological deterioration (Buljevac et al., 2002).

Bowel Dysfunction

Half of all patients need help with bowel management. A questionnaire survey of patients with SCI found that bowel dysfunction was a major problem, rated as only slightly less serious than loss of mobility (Glickman and Kamm, 1996). Bowel management may be equally problematic for patients with progressive spinal cord disease such as MS, with prevalence rates for bowel dysfunction reported at 30% to 50% (DasGupta and Fowler, 2003

Buy Membership for Neurology Category to continue reading. Learn more here