Nervous System Manifestations of Renal Disease



Nervous System Manifestations of Renal Disease


Yeong-Hau Howard Lien



Patients with chronic kidney disease (CKD) manifest a variety of neurologic disorders involving central, peripheral, and autonomic nervous systems. The severity of these nervous system manifestations increases in parallel to the advance of CKD. Without dialysis, patients with end-stage renal disease (ESRD) will develop uremic encephalopathy, uremic neuropathy, and uremic autonomic neuropathy. Some of these symptoms are partially or completely reversed by renal replacement therapy (i.e., dialysis or kidney transplantation). On the other hand, neurologic complications may occur due to improper hemodialysis, such as dialysis disequilibrium syndrome (DDS) and dialysis dementia, or due to complications of arteriovenous fistula (AVF) placement, such as ischemic monomelic neuropathy and vascular steal syndrome. In this chapter, CKD-associated neurologic disorders are reviewed in three sections: the central nervous system (CNS), the peripheral nervous system (PNS), and the autonomic nervous system (ANS).


CENTRAL NERVOUS SYSTEM

Several distinct CNS syndromes have been recognized in patients with ESRD: uremic encephalopathy,1 DDS,2 and dialysis dementia.3 In addition, dementia and cognition impairment, common in elderly populations, are significantly worsened by renal impairment, but poorly recognized in ESRD patients.4 These disorders are multifactorial and associated with prolonged hospitalization and an increased risk of mortality. Lastly, restless leg syndrome (RLS), a poorly understood syndrome, probably related to dopaminergic dysfunction in the subcortical system, affects 10% to 20% of ESRD patients5 and is discussed under the CNS section.


Uremic Encephalopathy


Manifestations

Uremic encephalopathy is an acute or subacute organic brain syndrome that occurs in patients with advanced renal failure and is frequently associated with GFR less than 10 mL/min/1.73 m2. The term uremic encephalopathy is used to describe the early appearance and dialysis responsiveness of the nonspecific neurologic symptoms of uremia. Patients with uremic encephalopathy display variable disorders of consciousness, psychomotor behavior, thinking, memory, speech, perception, and emotion.6,7 The symptoms may include sluggishness and easy fatigue; daytime drowsiness and insomnia with a tendency toward sleep inversion; inability to focus or sustain attention or to perform mental (cognitive) tasks and manipulation; inability to manage ideas and abstractions; slurring of speech; anorexia, nausea, and vomiting probably of central origin; imprecise memory; volatile emotionality and withdrawal; myoclonus and asterixis; paranoid thought content; disorientation and confusion with bizarre behavior; hallucinosis; transient pareses and aphasic episodes; coma; and convulsions.6,8

With an early recognition of CKD and the timely initiation of renal replacement therapy, severe uremic encephalopathy has been rare and is mainly related to acute kidney injury or unattended CKD due to a lack of health care. The severe neurologic symptoms of uremic encephalopathy such as seizure, confusion, myoclonus, and asterixis, are usually improved after a few runs of dialysis, and rarely recur if dialysis clearance is adequate. However, uremic encephalopathy may occur in patients on maintenance dialysis if they are not compliant with dialysis treatment, or if their dialysis prescriptions are not adequate. It should be mentioned that even with adequate dialysis, patients on maintenance dialysis may still have mild CNS symptoms, such as cognitive dysfunction as part of a “residual syndrome” because dialysis only replaces a fraction of total renal function.9


Pathogenesis

The mechanisms of uremic encephalopathy are multifactorial and largely unknown.1 It has been proposed that uremic encephalopathy is due to an accumulation of uremic toxins. Although many uremic toxins have been identified, exact toxins responsible for uremic encephalopathy are still unclear.10 Table 78.1 lists selected examples of uremic toxins grouped according to their structure. The source and characteristics of these toxins are provided.8









TABLE 78.1 Uremic Toxins8






























































Solute Group


Example


Source


Characteristics


Peptides and small proteins


β2-microglobulin


Shed from MHC


Poorly dialyzed because of large size


Guanidines


Guanidine, creatinine, guanidinosuccinic acid, methylguanidine


Arginine


Increased production in uremia


Phenols


p-Cresol sulfate


Phenylalanine, tyrosine


Protein bound, produced by gut bacteria


Indoles


Indican


Tryptophan


Protein bound, produced by gut bacteria


Aliphatic amines


Dimethylamine, trimethylamine


Choline


Large volume of distribution, produced by gut bacteria


Furans


CMPF


Unknown


Tightly protein bound


Polyols


Myoinositol


Dietary intake, cell synthesis from glucose


Normally degraded by the kidney rather than excreted


Nucleosides


Pseudouridine


tRNA


Most prominent of several altered RNA species


Dicarboxylic acids


Oxalate


Ascorbic acid


Formation of crystal deposits


Carbonyls


Glyoxal


Glycolytic intermediates


Reaction with proteins to form advanced glycation end products


MHC, major histocompatibility complex; CMPF, 3-carboxy-4-methyl-5-propyl-2-furanpropionic acid.


Modified from Meyer TW, Hostetter TH. Uremia. N Engl J Med. 2007;357:1316-1325.


Among them, uremic guanidino compounds, including creatinine, guanidine, guanidinosuccinic acid (GSA), and methylguanidine, are highly elevated in both the serum and cerebrospinal fluid (CSF) of uremic patients. GSA is a potential candidate as a uremic neurotoxin because it induces convulsion in animals at a dosage that produces a brain GSA level comparable to that of uremic patients.11 In vitro, GSA blocks both γ-aminobutyric acid (GABA) A receptors and glycine receptors. However, GSA-induced convulsions only respond to N-methyl-D-aspartate (NMDA) receptor antagonists. Further studies revealed that GSA causes neurotoxicity via activation of the NMDA receptor and by increasing Ca2+ influx.11 Whether these in vitro and in vivo findings have pathophysiologic significance in uremic encephalopathy is still controversial.

Parathyroid hormone (PTH) is another neurotoxin in a uremic state.1 PTH causes neurotoxicity by increasing intracellular Ca2+ concentration in brain cells. The total brain Ca2+ content is elevated in uremic patients and animals.1 The electroencephalogram (EEG) findings in uremic dogs are similar to those in patients with uremic encephalopathy. Both increased brain calcium content and EEG abnormalities in the uremic dog can be prevented by a parathyroidectomy.12 Furthermore, the intracellular Ca2+ level in brain synaptosomes is increased in uremic animals, which can be prevented by parathyroidectomy or reduced by verapamil, a calcium channel blocker.13 The increased intracellular Ca in uremic synaptosomes can be explained by an increased Ca2+ uptake and a decreased Ca2+ extrusion, both of which are mediated by PTH.13,14,15 Because calcium is an essential mediator of neurotransmitter release and a regulator of intracellular metabolic and enzymatic processes, alterations in brain calcium are likely to affect cerebral function.

Abnormal neurotransmitter content or release in the brain has been reported in uremic animals. Decreased brain norepinephrine content, uptake and release,16 and increased
acetylcholine content and release17 are found in uremic rats. In addition, the basal outflow of GABA and glutamate, but not the K+-stimulated outflow in the hypothalamus, which is measured by microdialysis, is increased in uremic rats. However, the K+-stimulated release of GABA is less sensitive to Ca depletion.18 Whether these changes in neurotransmitters in uremic rats are related to uremic encephalopathy in humans is not clear at present.



Prevention and Treatment

Uremic encephalopathy can be largely eliminated with the early referral of CKD patients to nephrologists and the timely initiation of dialysis. For patients on maintenance dialysis, the routine evaluation of dialysis adequacy and increased dialysis clearance for those who fall behind are effective in preventing uremic encephalopathy. As for treatment, intensive dialysis therapy—frequently, daily dialysis— should improve neurologic symptoms and signs within a week or two. A lack of response should prompt physicians to look for other etiologies. Although secondary hyperparathyroidism has been implicated in the pathogenesis of uremic encephalopathy, reducing the PTH level is rarely needed to improve neurologic symptoms. It is possible that repeated hemodialysis may reduce brain Ca21 content, thus reducing neurologic symptoms.1


Dialysis Disequilibrium Syndrome


Manifestations

DDS occurs rarely in current nephrology practice because of the early initiation of renal replacement therapy and routine orders of slow blood flow rate and short dialysis duration during the initial dialysis sessions. The cardinal symptoms of DDS are the symptoms caused by elevated intracranial pressure such as headache, nausea, and vomiting. These symptoms may progress into confusion, seizure, and even death if unrecognized and left untreated. The onset of DDS is usually during or immediately after aggressive hemodialysis, frequently in the setting of the first hemodialysis session.


Pathogenesis

The hallmark of DDS is brain edema that is induced by dialysis. Evidence accumulated that the “reverse urea effect” is the cause of DDS.2 Hemodialysis rapidly removes urea from the blood, but does not remove urea from the brain as efficiently. As a consequence, the urea concentration is higher in the brain than in the blood. This brain-blood urea gradient drives water to enter into brain tissue and causes brain edema and raises intracranial pressure. Elevated brain urea concentration and increased brain water content have been demonstrated in animals undergoing aggressive hemodialysis.28 When urea was added to the dialysate to keep the blood urea concentration equal to that in the brain, brain edema did not occur after hemodialysis. Using MRI, Galons et al.29 reported that the apparent diffusion coefficient of brain water increased in nephrectomized rats after hemodialysis. These results strongly suggest that the brain edema induced by hemodialysis in uremic rats is due to interstitial edema rather than cytotoxic edema, further supporting the reverse urea effect as the pathogenetic mechanism of brain edema in DDS. The increased diffusion coefficient of brain water after fast hemodialysis has been confirmed in patients with ESRD.20

More recently, a potential molecular basis for the reverse urea effect has been identified. In uremic rats, the brain expression of urea transporter 1 (UTB1) is reduced by 50%, whereas water channels aquaporin 4 (AQP4) and AQP9 were upregulated. Because of low UTB abundance, urea exit from the brain is likely delayed during the rapid removal of extracellular urea through fast dialysis. This creates an osmotic driving force that promotes water entry into the brain and subsequent brain swelling.30

There are other hypotheses for the pathogenesis of DDS, such as the creation of idiogenic osmoles, or paradoxical intracellular acidosis by hemodialysis.31,32 However,
Silva et al.28 measured most known organic osmoles in the brain after hemodialysis and did not find any significant changes in their concentrations. As for the acidosis hypothesis, it is not clear how intracellular acidosis causes interstitial edema. Although frequently quoted as the pathogenesis of DDS, one may wonder if cerebral acidosis is the consequence, rather than the cause, of brain edema in DDS.



Prevention and Treatment

It has been well established that limiting dialysis efficiency at the initiation of hemodialysis is the best way to prevent DDS (i.e., performing hemodialysis with a low blood flow rate, short dialysis time, and a small dialysis filter). This gentle dialysis treatment does not remove blood urea rapidly, thus preventing the formation of the brain-blood urea gradient.36 It is helpful to estimate the urea reduction rate from the dialysis prescription and target urea reduction to less than 60 mg per deciliter. Another common practice for preventing DDS is infusing mannitol during hemodialysis to raise serum osmolarity in order to reduce the brain-blood osmolarity gradient. The infusion of mannitol during hemodialysis is recommended for preventing DDS only in highrisk patients with marked azotemia (BUN level > 150 mg per deciliter) or in those with preexisting risk factors, as mentioned earlier. Mannitol infusion can cause acute volume expansion and congestive heart failure, and thus should not be used routinely for the initiation of hemodialysis.



Dialysis Dementia


Manifestation

This mysterious syndrome haunted dialysis patients in the 1970s, but it has now nearly vanished completely. Dialysis dementia, also named dialysis encephalopathy, is a progressive, frequently fatal neurologic disease almost appearing exclusively in patients being treated with chronic hemodialysis for more than 2 years. Early manifestations consist of a mixed dysarthria-apraxia of speech with slurring, stuttering, and hesitancy. Patients subsequently develop personality changes, including psychoses, paranoid thinking, or delirium, and global dementia, myoclonus, and seizures. In most cases, the disease progressed to death within 6 to 12 months.3 Observation studies revealed that dialysis dementia is a part of a multisystem disease that may include encephalopathy, osteomalacic bone disease, proximal myopathy, and anemia.3,38


Pathogenesis

Aluminum intoxication was first implicated in this disorder by Alfrey et al.39 Aluminum content of the brain’s gray matter, of bone, and of other soft tissue is markedly elevated in patients with dialysis dementia. Strong epidemiologic evidence links dialysis dementia to aluminum intoxication from the dialysate water and/or from oral phosphate binders containing aluminum.3 Furthermore, citrate was frequently used to correct metabolic acidosis in patients with advanced CKD. The gastrointestinal (GI) absorption of aluminum is markedly enhanced by the concomitant use of citrate.40 After a routine deionization of dialysate with reverse osmosis for removing contaminated aluminum in dialysis facilities and the limited use of aluminum containing phosphate binders, dialysis dementia has been nearly eliminated.



Prevention and Treatment

As mentioned earlier, dialysis dementia is nearly eliminated due to the use of reverse osmosis and the replacement of aluminum-containing phosphate binders with calcium-based binders or sevelamer. As mentioned earlier, citrate-containing alkylating agents enhance aluminum absorption and should not be used in conjunction with aluminum-based binders. As for treatment, DFO has been used for chelating aluminum. DFO is usually given at the end of hemodialysis. During the following dialysis session, the DFO-Al complex can be removed effectively by a polysulfone dialyzer.42 Aggravation of dialysis dementia by DFO may occur due to the release of aluminum from tissue.43


Dementia and Cognitive Impairment


Manifestations

With the marked reduction in the occurrence of uremic encephalopathy, DDS, and dialysis dementia, cognitive impairment and dementia have become the major CNS disorders in patients with ESRD. Dementia is characterized by a loss of function in multiple cognitive domains such as a decline in memory from previously higher levels of functioning, together with at least one of the following: aphasia, apraxia, agnosia, or disturbances in executive functioning. Cognitive impairment indicates that a patient’s ability to function in their work, personal, or social environment is affected.4 Both dementia and cognitive impairment are highly prevalent in patients with ESRD. Murray et al.44 reported that in 338 prevalent hemodialysis patients, 37% had severe cognitive impairment, qualified as dementia, 36% had moderate impairment, and 13% had mild impairment. In addition, the prevalence of cognitive impairment in ESRD patients increases with aging. It is 20% to 30% in patients 55 to 84 years of age, and increased to 50% to 60% in those 85 years or older.4

Cognitive impairment may already be present in patients with CKD stages 3 and 4. In the Heart Estrogen/Progestin Replacement (HERS) study, which involves 1,015 menopausal women with coronary artery disease, there is a fivefold risk of cognitive impairment if the estimated glomerular filtration rate (eGFR) is < 30 (stages 4 and 5) and patients with cognitive impairment tend to have a rapid progression of CKD.45 In the Reasons for Geographic and Racial Differences in Stroke (REGARDS) study46 involving over 23,000 adults > 45 years of age, it was found that eGFR < 60 is associated with a 23% increase in the prevalence of cognitive impairment after adjustment for demographic characteristics, prevalent cardiovascular disease, and cardiovascular (CV) risk factors.46 The prevalence of cognitive impairment increases in multiple areas with the decline of eGFR, as shown in Figure 78.2.47 For example, the prevalence of global cognition increases by 5% for each reduction of eGFR by 15 mL/min/1.73 m2. Other studies also confirm the association of CKD and cognitive impairment.46,48 In addition, the decline in cognitive function is faster in patients with CKD compared with non-CKD patients.49


Pathogenesis

There is strong evidence that indicates CKD-associated cognitive impairment is predominantly vascular in nature.
In the Cardiovascular Health Cognition (CVHS) study, it was found that moderate renal impairment in elderly adults (65 years or older) is associated with a 58% increase in the incidence of vascular dementia, but with no increase in the incidence of pure Alzheimer dementia. The overall incidence of vascular dementia and pure Alzheimer dementia in this cohort is 1.5% and 1.7% per year, respectively.50 In the Northern Manhattan Study (NOMAS), a prospective, community-based cohort of which a subset of stroke-free participants underwent MRIs, CKD stages 3 and 4 are associated with an increase in white matter hyperintensity volume, which is a marker for stroke, cognitive decline, and dementia.51 The increase of white matter lesions in CKD patients is confirmed by Ikram et al.52 More recently, Kobayashi et al.53 demonstrated that CKD also increases silent brain infarcts, and both the prevalence and the number of silent brain infarct increase with declining GFR.53 Furthermore, CKD is also associated with a rapid progression of carotid intima-media thickness in a community study.54






FIGURE 78.2 The unadjusted prevalence of cognitive impairment among 825 older adults (55 years or older) with mild-to-moderate renal insufficiency, according to estimated glomerular filtration rate (eGFR). Data taken from Yaffe K, Ackerson L, Kurella Tamura M, et al. Chronic Renal Insufficiency Cohort Investigators. Chronic kidney disease and cognitive function in older adults: findings from the chronic renal insufficiency cohort cognitive study. J Am Geriatr Soc. 2010;58:338-345. (See Color Plate.)






FIGURE 78.3 The proposed mechanisms of cognitive impairment and dementia in end-stage renal disease (ESRD). CKD, chronic kidney disease. (Modified from Kurella Tamura M,Yaffe K. Dementia and cognitive impairment in ESRD: diagnostic and therapeutic strategies. Kidney Int. 2010;79:14-22.)

The incidence of vascular dementia in ESRD patients is even higher than in those with moderate renal impairment. Fukunishi et al.55 reported that the 1-year incidence rate of vascular dementia and Alzheimer dementia is 3.7% and 0.5%, respectively, in aged Japanese hemodialysis patients. The incidence of vascular dementia was 7.4 times of that in the general elderly population. Furthermore, although in patients with moderate renal impairment the incidence of vascular and Alzheimer dementia is about the same, vascular dementia outgrows the Alzheimer dementia by sevenfold in ESRD patients.50,55

Therefore, it is likely that patients with CKD are at risk of vascular dementia and the progression of CKD is parallel to the progression of cerebrovascular disease— primarily, atherosclerotic disease. The pathogenesis of dementia and cognitive impairment in patients with ESRD is multifactorial, as illustrated in Figure 78.3. There are shared mechanisms for both CKD and neurovascular disorders, but nephrogenic and dialysis-associated mechanisms also play important roles in the development of dementia. The shared risk factors for CKD and cerebrovascular disease are aging, nonwhite race, low socioeconomic status/low education, diabetes, hypertension, hyperlipidemia.4 In addition, with declining renal function, several nephrogenic risk factors are likely to facilitate cerebrovascular disease, such as sympathetic overactivity, inflammation, oxidative stress, anemia, uremic toxins, and vascular calcification.4 Lastly, complications of hemodialysis such as intradialytic hypotension, hyperviscosity, thrombotic events, and hemorrhage due to heparin use will further worsen cerebrovascular disease.4,48




Prevention and Treatment

Because cognitive impairment and dementia associated with CKD are vascular in nature, reducing all cardiovascular risks before progressing to ESRD is critical for preventing these CNS disorders. Treatments targeted at controlling blood pressure, blood sugar, lipid profiles, proteinuria, and mineral metabolism may slow down the progression of both CKD and neurodegenerative diseases. Whether increasing dialysis clearance has a benefit on cognitive function has been controversial. Higher dialyzer urea clearance × time/urea volume of distribution (Kt/V) values have been associated with poorer cognitive function in patients on maintenance hemodialysis.44,57 However, in a small study with 12 patients, switching from conventional hemodialysis to nocturnal daily hemodialysis for 6 months resulted in a 22% reduction in cognitive symptoms, a 7% improvement in psychomotor efficiency and processing speed, and a 32% improvement in attention and working memory.58 Further studies are needed to substantiate these benefits from nocturnal hemodialysis.

Once dementia is diagnosed, the prognosis is poor in general because current drugs for treating dementia can only provide modest clinical benefit and none of them have been tested in patients with ESRD.4 The management of dementia associated with ESRD requires a multidisciplinary approach involving the primary care physician, caregiver, nephrologist, and staff at the dialysis facility and nursing facility to define the goals of care and to facilitate end-of-life care planning.


Restless Leg Syndrome


Manifestations

RLS is a common neurologic condition in patients with ESRD. The prevalence is 10% to 20% based on clinical diagnosis,5,59,60 but increases to 58% if diagnosed with a polysomnogram.61 RLS is characterized by an imperative need to move the leg because of uncomfortable and unpleasant sensations in the legs. It occurs primarily at rest, which is usually worse in the evening and alleviated by movement.


Pathogenesis

RLS is predominantly a disorder of the central rather than the peripheral nervous system. Dopaminergic dysfunction in the subcortical system appears to play a central role in idiopathic RLS. Reduced iron stores in the brain have been demonstrated, suggesting that the homeostatic control of iron is altered.62 Because iron is necessary for the activity of tyrosine hydroxylase, the rate-limiting step in dopamine synthesis, it is possible that a link exists between CNS iron deficiency and dopaminergic dysfunction. Compared with idiopathic RLS, RLS associated with ERSD progresses faster and responds poorly to a dopaminergic agent.63 Interestingly, iron deficiency seems to be linked to RLS in CKD patients with5 or without dialysis.64 Further studies are needed to define the role of disturbed iron homeostasis in the development of RLS in CKD patients. In addition, RLS tends to be exacerbated by caffeine, alcohol, and medications including dopamine antagonists, lithium, selective serotonin reuptake inhibitors, and tricyclic antidepressants.65

May 29, 2016 | Posted by in NEPHROLOGY | Comments Off on Nervous System Manifestations of Renal Disease

Full access? Get Clinical Tree

Get Clinical Tree app for offline access