Perspectives in Diabetes The Impact of Diabetes on the CNS ANTHONY L. McCALL

The brain is not usually thought to be a target of chronic diabetes complications. Nonetheless, substantial evidence, summarized herein, suggests that diabetes causes brain damage. Clinical syndromes of diabetes-related brain abnormalities are discussed along with possible causes. Various physiological effects of diabetes are reviewed, and questions are raised about gaps in our knowledge. Appropriate directions for future research are suggested. Diabetes 41:557-70, 1992

T

he impact of diabetes mellitus on the CNS has gained attention only recently (1-3). Peripheral neuropathy has been the primary neuroscience focus of diabetes research. Contrary to some early impressions, however, the CNS is not spared by diabetes (4). Chronically, diabetes mellitus afflicts the CNS in several ways. Diabetes increases stroke risk and damage, overtreatment with insulin or oral agents can permanently damage the brain, and diabetes may increase the prevalence of seizure disorders. Diabetes changes brain transport, blood flow and metabolism, and may produce a chronic encephalopathy. Acutely, glycemic extremes cause coma, seizures, focal neurological deficits, and impaired consciousness. The pathophysiological basis for these marked CNS abnormalities seen in hypoglycemia, hyperosmolar coma, and ketoacidosis are largely unknown. The purpose of this article is to'highlight some of the

From the Diabetes Program, Department of Veterans Affairs Medical Center, Portland; and the Departments of Medicine, Neurology, and Cell Biology and Anatomy, Oregon Health Sciences University, Portland, Oregon. Address correspondence and reprint requests to Dr. Anthony L. McCall, Director, Diabetes Program, Medicine (111-P), Portland VA Medical Center, Portland, OR 97207. Received for publication 5 December 1991 and accepted 27 December 1991.

DIABETES, VOL. 41, MAY 1992

brain abnormalities identified in diabetes mellitus, raise questions about the extent of our knowledge of diabetes and the brain, and draw attention to potentially important future research in relevant interdisciplinary areas. The major focus will be the chronic effects of diabetes on the brain. DIABETES MELLITUS AND STROKE Diabetes increases stroke risk and damage—epidemi-

ology. Epidemiological studies demonstrate that diabetes mellitus carries a two- to sixfold increased risk of thrombotic (but not hemorrhagic) stroke. Diabetes is believed to cause 7% of deaths due to stroke, and cerebrovascular disease may be present in 25% of patients dying with diabetes (5). Because non-insulindependent diabetes mellitus (NIDDM) is underdiagnosed, estimates of the contribution of diabetes to stroke risk may be low. Diabetes increases risk of atherosclerosis in all vascular beds including the brain. Hypertension, a major stroke risk factor, is more common in those with diabetes. However, the increased risk of stroke in diabetic individuals persists even when we correct for other concomitant risk factors, i.e., hypertension, that occur more commonly with diabetes mellitus (6). One noteworthy at-risk group is elderly women. In the Framingham Study, the incidence of atherothrombotic brain infarction rose from ~50/10,000 per yr in nondiabetic women 70-79 yr old to 150/10,000 per yr in diabetic women 70-79 yr old (5). Elderly women represent an increasing proportion of our population. Even modest hyperglycemia increases the risk of stroke significantly. The relative risk of stroke in men increases by approximately threefold when their plasma glucose 1 h, after a 50-g glucose load, rises from 10%) comparable to treated diabetic patients, in 50 patients with stroke or transient ischemic attack (TIA). Excluding known glucose intolerant or diabetic patients, there were 21 of 50 stroke or TIA patients with apparently unrecognized diabetes mellitus. Confirming these data, in a prospective study of 200 acute stroke patients, Gray et al. (17) found that 27% had an HbA! >2SD above the laboratory mean without a history of diabetes. A final, very important possibility is that hyperglycemia predicts excessive stroke morbidity and mortality because glucose acts directly or indirectly to exacerbate brain damage during ischemia. Several human studies have suggested that stroke outcome and severity of residual neurological deficits may be predicted by the glucose concentration in the plasma at the time of stroke and that the damage is due to hyperglycemia per se (11-13,17). The concept of glucose as a potential brain toxin originates largely from animal studies (14). HYPERGLYCEMIA WORSENS ISCHEMIC BRAIN DAMAGE IN ANIMAL MODELS Acute hyperglycemia (usually produced by i.p. injection of glucose) before experimental global or forebrain ischemia (types of brain ischemia similar to that observed after cardiac arrest) increases histological damage to the brain and worsens neurological outcome (14,18-34). In contrast, acute hypoglycemia protects against such damage. Hyperglycemia subsequent to stroke seems less important to neurological morbidity in experimental ischemia; glucose administration and hyperglycemia after the ischemia have little impact. Such findings raise serious questions about whether glucose administration should be omitted routinely from resuscitative measures (35). Fewer studies address whether chronic diabetes in animal models worsens stroke damage. However, those

558

that do suggest that brain damage from ischemia is worsened by diabetes (14,36,37). Because damage occurs in the absence of atherosclerosis, these studies implicate hyperglycemia as a possible cause of excessive damage. The relationship of stroke damage and hyperglycemia focuses attention on understanding brain ischemia in metabolic terms and raises the possibility of pharmacological remedies to block critical metabolic abnormalities during ischemia. In particular, aspects of glucose metabolism during ischemia are important for two major pathways thought to be relevant to brain ischemia—the toxic accumulation of metabolic acid and the toxic overstimulation of neurons by excitatory amino acids, glutamate, and aspartate. ACIDOTOXIC BRAIN DAMAGE AND PANNECROSIS The mechanism usually imputed to explain the increased stroke damage after hyperglycemia is cellular acidosis (19,21,38). This "acidotoxicity" hypothesis states that anaerobic metabolism of glucose to lactic acid under conditions of oxygen lack leads to intra- and extracellular acidosis. Indeed, cellular pH after ischemia is a direct function of lactic acid levels in brain. The acidity predicts pannecrosis in the brain, i.e., cellular damage not only to neurons but also to glia and vascular elements. Evidence suggests both pH and lactate levels may independently contribute to cellular damage (39). How lactate or low intra- and extracellular pH cause the damage is not known but will be the focus of much future work. Even a difference of a few millimolars in blood glucose seen after a few days of dexamethasone pretreatment has been invoked to explain more severe stroke damage in rats (20). A major question to be addressed is how such modest hyperglycemia might exert a major effect on stroke outcome (40). Could small increments in glycemia markedly increase lactate-induced brain injury and is this consistent with a mass-action effect? Possibly some other variable or variables serve to amplify the glucose effect. One explanation is that altered glucose transport at the blood-brain barrier or at the plasma membrane of neurons and glia (see below) could serve as such an amplifier, augmenting lactate accumulation during ischemia by increasing glucose delivery to brain. Any theory relating glycemia to stroke damage must explain observations suggesting both possible detriment and benefit of glucose in ischemic brain injury. In animal models of focal brain ischemia (like that seen after thrombotic stroke), there is evidence for worsening (34,41) and some sign of improvement (42) in cellular damage by hyperglycemia. In an area of dense ischemia (the center of an infarct), glucose typically worsens damage. In the penumbra, the ischemic zone that surrounds dense infarction, however, glucose may ameliorate damage. In hypoxia, hyperglycemia may improve brain damage (43). A major question is what relevance data from these animal models have to different types of human cerebral ischemia. Why does hyperglycemia worsen some types of isch-

DIABETES, VOL. 41, MAY 1992

A.L. McCALL

emic/hypoxic brain damage but actually improve others (41)? Inefficient ATP generation from glycolysis and toxic buildup of cellular lactate in areas of severe oxygen lack may be the answer. Glycolysis generates a net yield of 2 ATP equivalents compared with as much as 38 for complete glucose oxidation. In addition, a block of pyruvate dehydrogenase characterizes some forms of ischemia (23) and could accelerate cellular accumulation of lactic acid and acidotoxicity. In less hypoxic areas by contrast, more efficient oxidative energy generation and less lactate accumulation could occur. Glucose may benefit the heart, enhancing brain survival by improving cardiac output. Research efforts are needed to address these possibilities. In addition, more studies of how chronic diabetes or glucose intolerance exacerbates neurological damage in animal models of stroke (both focal and global) are needed. Specifically, we would like more information about the dose-response characteristics of the relation between ischemic brain damage and degrees of hyperglycemia and the effect of diabetes treatment on damage. The clinical implications of possible glucose toxicity to brain during stroke are several. If hyperglycemia is implicated in human stroke damage and does not simply reflect unrecognized diabetes or stress hyperglycemia from severe infarction, then what should be done? How tightly controlled should glycemia be in diabetic stroke patients? Does the answer depend on the type of stroke as suggested by Helgason (14)? Should we maintain tight glucose control, potentially risking hypoglycemia in acute stroke patients? What is appropriate nutritional therapy for diabetic stroke patients who can not feed themselves? Should glucose be omitted from their intravenous therapy or should very-low-glucose tube feedings be used when needed? There are no answers to these questions. Clinical studies should address these issues.

EXCITOTOXIC BRAIN DAMAGE

A second type of brain damage occurs during ischemia (and hypoxia or hypoglycemia)—excitotoxicity from acidic amino acids. The characteristic histopathology shows selective neuronal necrosis, with particular neurons in specific brain regions affected but sparing of glial and vascular elements. Biochemical correlates have been sought to explain the selective, regional nature of this brain damage. Most of the focus has been on glutamate toxicity via specific receptors, particularly the A/-methyl-D-aspartate (NMDA) receptor (44), although other receptor types are also involved (45). This receptor, one of the major types of brain glutamate receptors, becomes overstimulated by the release of glutamate from presynaptic neurons during conditions of energy deprivation in the brain. Membrane depolarization occurs, ionic movements follow, and intracellular calcium particularly increases. One attraction of the NMDA toxicity hypothesis is its potential to explain selective neuronal vulnerability. Selective vulnerability should occur where NMDA or other involved glutamate receptors are located postsynapti-

DIABETES, VOL. 41, MAY 1992

cally. A second appeal is the potential to intervene experimentally with drugs that block the receptors. It is likely, based on in vitro studies of glutamate toxicity in cultured cerebellar neurons, that glucose metabolism is intimately involved in generating the energy locally that determines whether NMDA toxicity occurs (46,47). In particular, glucose oxidation may unblock the magnesium channel in the NMDA receptor. During ischemia or hypoglycemia, glutamate may literally stimulate some neurons to death. Normally, glutamate release would simply cause postsynaptic neurons to receive an excitatory postsynaptic potential and possibly depolarize as a result. Under conditions of energy failure, however, i.e., oxygen or glucose lack, excessive release may occur, with a concomitant failure of the energy-dependent reuptake mechanisms that detoxify glutamate. The result is hyperstimulation of the postsynaptic neuron, a toxic event made worse by inability to restore ionic homeostasis due to the original energy failure. One characteristic consequence of these events is intracellular calcium overload in particular neurons. Cellular calcium overload represents another important biochemical event during ischemia, hypoglycemia, and hypoxia (48). As with the other aspects of excitotoxicity, it may explain the selective nature of neuronal damage. It also offers the possibility of pharmacological intervention to ameliorate the damage. There may be a connection between these different metabolic mechanisms of brain damage. Nimodipine, a calcium-channel blocker, mitigates the acidosis and possibly brain damage caused by hyperglycemia during experimental ischemia (49).

HYPOGLYCEMIA AND BRAIN DAMAGE

Human studies. Hypoglycemia depresses brain metabolism. Based on studies by Kety et al. (50), it became clear that insulin coma, deliberately induced as a therapy for mental disorders (a practice laudably abjured), markedly reduces cerebral oxygen consumption and disproportionately decreases brain glucose metabolism. Hypoglycemia can produce permanent brain damage and death. The cortex, caudate, and hippocampus appear most vulnerable to hypoglycemia. However, confounding variables mar interpretation of some of the human case studies (51). Overall, the brain in humans dying of hypoglycemia may appear either grossly normal or markedly affected, depending on whether the clinical course has been complicated by repeated seizures and cardiac arrest (51,52). Cortical and hippocampal atrophy and ventricular enlargement may occur in long-term survivors. Laminar necrosis occurs in the cerebral cortex, especially in the third and fifth layers. Sometimes, diffuse demyelination is observed. Certain zones of the hippocampus may be characteristically involved. The striatum can be necrotic but involvement of the globus pallidus is uncommon. The cerebellum is relatively spared, especially the Purkinje cells. Animal literature. Animal studies of acute hypoglycemic brain damage, particularly those of Brierley et al. (53) and Auer et al. (56), gave a more controlled examination of histopathological effects of hypoglycemia on the brain.

559

DIABETES AND CNS

Brierley and associates (53,54) argue for the similarity of hypoglycemic brain damage to ischemia and hypoxia, whereas Auer et al. emphasize the differences. Two major types of histological change are observed in hypoglycemia-induced brain damage (55). Cellular shrinkage and condensation of nuclei and cytoplasm and scalloping of the cytoplasmic membrane occur. A second type of histological neuronal injury is swollen neurons, often with vacuolization (probably swollen mitochondria) seen near the cytoplasmic membrane. In any case, selective neuronal necrosis is the most characteristic brain lesion of hypoglycemia. Based on extensive studies in rats with acute hypoglycemia, Auer et al. (56-62) concluded that recognizable neuronal injury leading to cellular death takes place within 2-8 h after hypoglycemia severe enough to produce an isoelectric electroencephalogram (EEG). Cells in the cortex, hippocampus, and caudate are involved in lesions first visible in the dendrites but that comparatively spare axons.

NEUROCHEMICAL EFFECTS OF HYPOGLYCEMIA Acute hypoglycemia produces cerebral energy failure, causes loss of ion homeostasis (especially increases in intracellular calcium) (19,48,63-66), produces membrane depolarization, alters intermediary and amino acid metabolism, depresses protein synthesis (67), increases phospholipid hydrolysis and concentrations of free fatty acids especially arachidonate, and alters cyclic nucleotide metabolism. Moderate hypoglycemia produces a partial energy failure of the CNS through brain fuel starvation or neuroglycopenia, clinically manifest at ~3-mM glucose concn (68-70,214-216). One consequence of the energy failure is synaptic release of glutamate (or aspartate) and a concomitant failure of energy-dependent reuptake of excitatory neurotransmitters. Excitotoxic cellular damage, as with stroke, is mediated via specific receptors, particularly the NMDA receptor (see above). Hypoglycemic brain damage may be prevented by blocking the NMDA receptor. Thus, Wieloch (71) showed in rats that the experimental compound, AP7, inhibits binding of NMDA to its receptor and can prevent almost 90% of the histological damage of hypoglycemia associated with an isoelectric EEG. Auer et al. (72) showed similar protection with a different NMDA blocker (72). Siesjo et al. (48,65,66,73,74) suggested that calciummediated brain damage occurs in hypoglycemia and other brain insults, such as stroke. Selective vulnerability to brain damage may occur because a greater density of calcium channels on some cells permits greater cytoplasmic calcium influx. Intracellular calcium overload activates proteases and lipases, exerting toxic effects through reactions catalyzed by these enzymes and causing mitochondrial swelling and dysfunction. Calcium toxicity may be blocked pharmacologically and potentially prevent the neurotoxic consequences of hypoglycemia (and stroke) to the brain. Thus, even if serious hypoglycemia cannot be avoided, its associated brain damage could be minimized.

560

SOME UNANSWERED QUESTIONS Several questions emerge from a review of the neuroscience literature on hypoglycemia. What relationship exists between recurrent clinical hypoglycemia, e.g., repeated bouts of coma or seizures but with apparent neurologic recovery, and long-term brain damage? Frier et al. (75, 76) and others found that repeated hypoglycemia predicts cumulative brain damage (75,76). Do mild bouts of hypoglycemia produce selective neuronal necrosis as in rat studies (56)? If such pathology does not occur, is there some other form of sublethal (e.g., metabolic [77]) neuronal (or glial) injury that impairs neurological function in the long run? Are the brain consequences of hypoglycemia preventable by calcium channel blockers or NMDA receptor blockers? Do some areas of the brain fail to adapt to repeated hypoglycemia (78), although animal studies suggest that much of the brain appears to adapt? What is the role of CNS in counterregulatory hormone defects observed with repeated hypoglycemia (79), and if there is a significant CNS role, what is its neurochemical basis? Is it time for us to design clinical studies in high-risk patients based on our knowledge of the neurochemistry of hypoglycemia in animals? Certain studies, e.g., the use of calcium channel blockers in patients with intensively treated diabetes and hypertension to protect the brain from hypoglycemic damage, seem ethically appropriate. A last question, best addressed in experimental animals, is whether the neurochemistry of hypoglycemia differs in the context of preexisting diabetes. BRAIN TRANSPORT, BLOOD FLOW, AND METABOLISM IN DIABETES AND HYPOGLYCEMIA Transport of glucose and other nutrients into the

brain. Glucose transport into the brain of diabetic animals appears to be depressed as a direct or indirect consequence of hyperglycemia (80-85). With different techniques, several groups have independently reported depressed glucose transport across the blood-brain barrier in experimental diabetes. Early studies by Ruderman et al. (86) found, with cerebral arteriovenous differences, a method influenced by both transport and metabolism, that the net extraction of glucose by the brain is decreased by one-third in uncontrolled diabetes. With the intravenous infusion method to study unidirectional flux of glucose into the brain (a true index of transport), Gjedde and Crone (87) found that chronic hyperglycemia decreased the blood-brain barrier transport of glucose from 44 to 25%. A decreased maximum transport capacity (Tmax) for glucose from 4 to 2.9 ixmol • g~1 • min~1 explains this decrease. When plasma glucose in diabetic rats is lowered to normal values, the glucose transport rate into brain is 20% below that of nondiabetic rats. In extensive physiological studies in diabetic rats, we (81,82) used the Brain Uptake Index method to study transport of several hexoses and other metabolic substrates. Glucose transport into the brain decreases from 40% (control) to 27% (diabetes) in a single cerebral circulation passage. As previously shown (87), the maximal transport rate decreased (from 1.8 to 1.0 ^mol • g~ 1

DIABETES, VOL. 41, MAY 1992

A.L McCALL

• min~1). Hexose transport is symmetrically depressed on both sides of brain capillaries; both efflux and influx of the nonmetabolized hexose analog, 3-O-methyl-D-glucose, are decreased to a similar degree (82). Transport of hexoses into the brain in diabetes is generally but specifically altered. Transport of other nonglucose sugars is depressed by diabetes, whereas that for the ketone body, fj-hydroxybutyrate, is increased in untreated diabetic rats but depressed after insulin treatment (82). Transport of other substrates, e.g., amino acids, or water is unaffected. Chronic changes in antecedent glycemia appear to be the primary stimulus for altered glucose transport into the brain. This conclusion is based on studies showing that glucose transport and glycemia correlate with insulin treatment, sucrose feeding plus insulin treatment, and starvation of diabetic rats (82). The time course of these changes in blood-brain barrier glucose transport is slow, requiring >1 day in rats. Recently, Pardridge et al. (83) confirmed and extended findings on the decrease in blood-brain barrier glucose transport in experimental diabetes mellitus. Choi et al. (84) found increased GLUT1 mRNA levels in the brain capillaries of rats with streptozocin (STZ)-induced diabetes but decreased transport, with the in situ brain perfusion (88) method, and decreased levels of GLUT1 protein (the major blood-brain barrier glucose transporter) in the same brain capillaries (83). They (84) found a 44% decrease in glucose transport and a 77% decrease in GLUT1 immunoreactivity (based on a COOH-terminal and presumably specific antiserum) in isolated cerebral microvessels from rats with experimental diabetes (84). Mooradian and Morin (85) also confirmed the decrease in blood-brain barrier transport but found no change in the number of glucose transport proteins in brain capillaries from diabetic rats. Some controversy has arisen over whether the changes in blood-brain barrier glucose transport are specific or indeed even exist. Harik et al. (89-91) found decreased transport of glucose into the brains of both acutely and chronically hyperglycemic rats but explained the results on the basis of altered cerebral blood flow and/or altered diffusion of L-glucose, their control marker for nonspecific transport. Duckrow (92) found that no transport change exists for glucose at the blood-brain barrier in diabetic rats, whereas, in another study (93), he described a slight decrease. Pelligrino et al. (94) argued against a decrease in glucose transport across the blood-brain barrier in diabetes but found strong direct evidence for increased blood-brain barrier transport in chronic hypoglycemia. Pardridge et al. (83) argued that the methods used in studies failing to find decreased glucose transport in diabetes examine glucose transport into the brain at levels of glucose that largely, if not completely, saturate the carrier proteins. Thus, they would not be able to demonstrate any significant or consistent transport differences. In any case, some controversy will continue, at least until studies are conducted to assess adequately whether such effects occur in humans. In addition, there needs to be more basic information about where glucose

DIABETES, VOL. 41, MAY 1992

transport proteins are distributed in the brain and how they are regulated. In particular, it seems especially likely that different glucose transport proteins will be differentially regulated.

BRAIN TRANSPORT IN CHRONIC HYPOGLYCEMIA Chronic hypoglycemia increases glucose transport into the brain. Chronic but not acute hypoglycemia increases blood-brain barrier transport of glucose (96). Rats made chronically hypoglycemic by insulinoma implantation, insulin infusion with an osmotic minipump, or daily injections of PZI insulin all have increased brain glucose transport. As in diabetic rats, the changes in blood-tobrain transport in chronic hypoglycemia are specific and rather slow in their onset. Other transport systems are either unaffected (e.g., large neutral amino acids or choline transport) or affected in an opposite fashion (e.g., lactate and pyruvate transport.in insulinoma rats). Furthermore, glucose-6-phosphate, creatine- phosphate, and ATP levels in brain are not significantly lower in insulinoma rats, suggesting the increased glucose extraction at the blood-brain barrier protects brain energy metabolism. Pelligrino et al. (63, 64) similarly found evidence for an increase in blood-to-brain transport of glucose in chronically hypoglycemic rats (78). In these studies, regional analysis suggested that most, although not all, brain areas adapt. The effect of these adaptations are to enhance the cerebral metabolism of glucose. However, note that in chronic hypoglycemia there is less compensatory cerebral blood flow change (97) and, despite adaptation in glucose transport, abnormal mitochondrial respiration remains (77). It appears from the above studies that hyperglycemia and hypoglycemia chronically decrease and increase, respectively, transport of glucose at the blood-brain barrier. Possibly, glycemia similarly affects the cytoplasmic membrane barrier of brain parenchymal cells, particularly glial cells (98,99). Glucose starvation of many tissues and cell types has been repeatedly shown to increase glucose transport and conversely, glucose exposure to decrease glucose transport (100,101). It seems likely that the brain behaves similarly, based on studies performed to date. Regarding controversies in studies of brain transport in diabetes, although we might believe that analysis of glucose transport proteins and mRNA will provide a definitive answer, this may not be. Molecular biology analysis is helpful insofar as it confirms and explains how transport changes. Studies of transport must stand or fall on their own merits. Transport of other substrates and ions in diabetes. Mooradian (102) found that the maximal rate of choline transport across the blood-brain barrier declined from 2.2 to 0.14 nmol • min~1 • g~ 1 in chronically diabetic rats. Insulin therapy for 5 days did not rectify the abnormal choline transport. Because choline may be an important precursor to brain acetylcholine, a neurotransmitter thought to be involved in age- and Alzheimer's diseaserelated memory loss (103), this is an intriguing finding. Mooradian (102) also found that dehydroascorbate,

561

DIABETES AND CNS

but not ascorbate, in vitro was able to compete with glucose for entry into several tissues including the brain. However, it is not clear whether hyperglycemia could cause a deficiency of this vitamin in the brain and lead to dysfunction as a result. Specific abnormalities in sodium transport at the blood-brain barrier in experimental diabetes were observed (104-107). The permeability surface area product (PS) for sodium was determined by infusing 24 Na + i.v. for up to 15 min in diabetic and control rats. The PS decreased from 7.1 in the frontal cortex of control rats to 5.4 x 10~5 • cm" 3 • g " 1 • s" 1 after 2 wk of STZ-induced diabetes (104). Similar decreases occur in occipital cortex after 2 wk of diabetes mellitus. 36CI- and 3 Hsucrose transport do not differ significantly between diabetic and control rats (104), however. Potassium permeability (107) also decreases by 39%, but no difference exists in calcium permeation into the brain. Insulin treatment restores Na+ transport to near-normal values within a few hours (105). Transport changes are not explained by hyperosmolarity because 50% mannitol increases Na+ transport, myo-lnositol treatment normalizes the deficient Na+ transport. It has been postulated that abnormal Na+-K+-ATPase activity in cerebral endothelial cells is the cause of these effects (106). Abnormal permeation of protein into the brains of animals with experimental diabetes mellitus also may occur. Stauber et al. (108) found a selective extravasation of albumin but not IgG or complement (C3) in immunohistochemical studies of the brain in STZ—induced diabetes of 2 wk duration. However, studies by Williamson (109) did not find that 125l-labeled albumin permeation into brain is increased in animal diabetes. Perhaps the latter method is less sensitive than immunohistochemistry. If very poor diabetic control affects microvascular permeability to albumin or sodium of the blood-brain barrier, it could be relevant to the syndrome of cerebral edema that has been observed, particularly in children being treated for diabetic ketoacidosis (110— 114). CEREBRAL BLOOD FLOW AND VASCULAR REACTIVITY CBF probably decreases in animals with poorly controlled, hyperglycemic diabetes (83,89,115-119). Some studies are contradictory (120), perhaps due to experimental design issues such as measurement technique and diabetes duration and severity. CBF may be depressed in a moderate, regionally specific fashion, which depends on the severity of hyperglycemia. Particularly affected is the hindbrain (14% reduction at glucose of 29 mM), with less effect on the forebrain. CBF reduction has been seen in rats with either acute hyperglycemia from intraperitoneal glucose or STZ-induced chronic diabetes (115). It may not be simply an osmotic effect because equiosmotic mannitol yields a more modest decrease. Because the risk and severity of stroke in diabetes is increased, such findings may be mechanistically relevant. If the findings apply to humans with poorly controlled diabetes, they might lead to critically diminished blood flow in certain brain areas, particularly in the presence of extracranial atherosclerosis or thrombosis.

562

However, it is not certain whether good-to-moderate glycemic control in people with diabetes produces similarly altered CBF as occurs in these animal models. Some studies find decreased basal CBF in diabetes (121). However, some studies have failed to find such changes or actually found increases in CBF in diabetic humans (122-126), possibly because people are not usually studied in very poor glycemic control. Furthermore, even if CBF is normal under basal conditions, cerebrovascular reactivity is often abnormal in diabetes. Cerebrovascular reactivity may be selectively impaired in long-term experimental and human diabetes (116,120, 127,128). For example, Pelligrino and Albrecht (116) examined CBF responses to hypoglycemia, hypoxia, and hypercarbia after either 6-8 wk or 4 - 6 mo of STZinduced diabetes in rats. In these studies, basal CBF is decreased and vascular reactivity (dilation and increased flow) blunted. Hypoglycemia-induced increases in CBF in long-term diabetic rats are severely impaired. Similarly, human studies of cerebrovascular reactivity, both of dilatation and constriction show abnormal responses (123-126,129,130). The duration of diabetes may be important because not all studies confirm these results. Abnormal cerebrovascular reactivity in people with long-standing diabetes may be specific to certain stimuli (130). There is evidence that clinical diabetic neuropathy predicts the presence of abnormal cerebral vascular reactivity. What is the import of abnormal vascular reactivity in response to hypoglycemia? Increased CBF during hypoglycemia is thought to enhance the supply of glucose to the brain. Impairment of vascular reactivity may blunt this compensatory increase. Cohen et al. (131) obtained evidence for a vascular neuropathy in the carotid arteries of alloxan (ALX)-induced diabetic rabbits (131). Norepinephrine content, release, and uptake are all abnormal in this model. It is not known whether smaller resistance vessels are similarly affected, although evidence exists for their noradrenergic innervation. These investigators also found abnormal cholinergic relaxation in large blood vessels in experimental diabetes, with evidence pointing to abnormal vasoconstrictor prostanoid release as a mechanism (132,133). Although these studies were not performed in brain blood vessels, note that abnormal prostanoid metabolism was found in diabetic brain (134), although not in diabetic brain microvessels (135). DIABETES AND HYPOGLYCEMIA AFFECT MICROVASCULAR METABOLISM IN THE BRAIN The cerebral microvasculature may be a target for chronic diabetic complications. Pericyte loss in brain capillaries was observed in animal models of diabetes (136). However, the effects of diabetes on pericytes in brain capillaries is less well established than effects in retinal capillaries. Reske-Nielsen et al. (137) described brain vascular abnormalities in long-term insulin-dependent diabetes mellitus (IDDM) patients. In addition, in autopsies of humans with IDDM, there is microvascular pathology present including basement membrane thickening (138). Metabolic abnormalities, particularly in energy metab-

DIABETES, VOL. 41, MAY 1992

A.L McCALL

olism (3,14,36,37,139,140), exist in brain microvessels from animals with experimental diabetes. Frequently, altered metabolism of a fuel in vitro by rat brain microvessels correlates with the transport of that fuel in vivo across the blood-brain barrier (82,96,139,140). Thus, diabetes depresses the metabolism of glucose in brain microvessels from rats with STZ-induced diabetes. Microvessel oxidation of glucose decreases by 65-83% and conversion to lactate by 21-61%. As with glucose transport in vivo, prior glycemia is an important regulatory factor for microvessel metabolism. Restoration of euglycemia by insulin treatment or starvation for days both lowers blood glucose levels and rectifies microvessel glucose metabolism. In our studies, [1-14C]lactate oxidation was depressed in brain microvessels from diabetic rats, suggesting impaired flux through the pyruvate dehydrogenase pathway. Restoration of glycemic control improved flux through this pathway. Hingorani and Brecher (141) confirmed and extended work on the abnormal fuel metabolism of cerebral microvessels from diabetic animals in their studies of New Zealand rabbits with 150 mg/kg ALX-induced diabetes. They studied isotopic oxidation of [6-14C]glucose or [1-14C]oleate and the incorporation of radioactivity into CO2, lactate, triglycerides, cholesterol ester, and phospholipids. They found competition between the oxidation of glucose and oleate. Thus, incubation of microvessels with 5.5 mM glucose reduces oleate oxidation by 50%. Conversely, glucose oxidation is reduced by incubation with oleate. Our laboratory found that the oxidation of glucose and p-hydroxybutyrate is altered in a coordinated but opposite direction in brain microvessels of rats with diabetes and chronic hypoglycemia (due to a subcutaneous insulinoma) (139). Glucose oxidation by isolated cerebral microvessels is depressed in diabetes but increased in chronic hypoglycemia, paralleling the transport of glucose in vivo across brain capillaries (82,96). Acutely, insulin treatment has no effect on microvessel metabolism. Oxidation of p-hydroxybutyrate to CO2 by isolated cerebral microvessels is increased in diabetes, as is its transport into brain, whereas, in chronic hypoglycemia, depressed oxidation occurs. The basis for these parallels between fuel transport and metabolism is unknown. It seems implausible that glucose transport by microvessels limits their metabolism because the transport must be abundant enough to provide glucose for the whole brain (151). On the other hand, local (i.e., capillary) metabolism may well influence transport, a notion with considerable precedent in studies of glucose starvation and refeeding in fibroblasts (100,101,142-148). Precise knowledge of the metabolic control mechanisms that regulate transport may in the future permit their deliberate manipulation as therapy. Altered fuel metabolism by brain capillaries does not affect their ATP content or ATP-ADP ratios after 1 or 8 wk of diabetes in rats (139). Although ATP remains unchanged, oxygen consumption by microvessels from diabetic rats increases by 35% compared with controls. The specific metabolic pathways that explain the increased oxidative metabolism are unknown. Possibly, ion

DIABETES, VOL. 41, MAY 1992

homeostasis in the brain microvasculature of diabetic animals requires a higher rate of respiration. In poorly controlled diabetes mellitus combined with situations of fuel deprivation, e.g., hypoxia or ischemia, excessive oxygen consumption or other abnormalities of fuel use may produce microvascular dysfunction in brain. Future studies should be directed toward looking at the functional consequences of this abnormal fuel metabolism. A major question to be addressed is why there is no obvious clinical syndrome equivalent to diabetic retinopathy that affects the brain, e.g., with hemorrhages. Pericyte density is one possible answer. Retinal capillaries have about four times as many capillary pericytes as the brain. These cells may be a particular target of diabetic microvascular damage. What are the mechanisms involved in CBF changes? No answer is available. If hyperglycemia causes the CBF changes, good control may prevent them. BRAIN ENERGY METABOLISM Kety et al. (149) examined the effects of diabetic coma (ketoacidosis) on brain metabolism in humans. Cerebral metabolic oxygen consumption was reduced by 50%. This reduction probably represents a decrease related to the coma rather than a specific effect of diabetes. Other causes of coma, hepatic failure, uremia, and anesthesia, all produce similar decreases in brain oxygen consumption (150). Ruderman et al. (86) showed that brain fuel metabolism is altered in experimental diabetes. As in starvation, during ketotic diabetes in animals, the brain slowly decreases its utilization of glucose and increases its utilization of the alternate fuels, the ketone bodies, acetoacetate and (3-hydroxybutyrate. This occurs in both anesthetized and unanesthetized rats (152). With arteriovenous differences (86) and sampling of intermediary metabolites in the brain, it was possible to show a glucose-fatty acid cycle in brain during uncontrolled diabetes (153). The effects of poorly controlled diabetes on specific pathways of brain energy metabolism are complex (86,154-156). It seems likely that ketosis engenders a switch from brain utilization of glucose to ketone bodies, much as has been observed in starvation (155,157). Animal and human studies (93,94,155,158-160) have been conflicting on the extent to which nonketotic diabetes influences cerebral metabolism. In humans with well-controlled IDDM, Grill et al. (122) has performed studies of regional CBF and metabolism before and after insulin normalization of glycemia. Before insulin treatment, an increased arteriovenous difference for ketone bodies was seen, as in animal studies. Cerebral nonoxidative metabolism of glucose was increased in IDDM (19 vs. 7% in nondiabetic subjects), and the ratio of glucose to oxygen consumed was altered. The fate of the extra glucose is unknown. Also, which fuel substitutes for glucose oxidation is uncertain. Studies from Jakobsen et al. (117) in diabetic rats suggest that both an increase and then a decrease sequentially in cerebral glucose metabolism may be observed, depending on the time

563

DIABETES AND CNS

course of diabetes (and possibly hyperglycemic severity). More studies need to be done, especially in NIDDM patients. BRAIN NEUROTRANSMITTER METABOLISM Experimental studies indicate that binding and metabolism of neurotransmitters within the brain, especially monoamines, are abnormal in untreated diabetes mellitus. Dopaminergic neurotransmission may be diminished. Lozovsky et al. (161) reported that the number of dopamine receptors, measured by 3H-spiperone binding, is increased by 30-35% in ALX- or STZ-induced diabetic rats. Dopamine receptor ligand binding is normal in diabetic rats treated with insulin, but dopamine metabolism may not be entirely normalized after insulin therapy (162). Similar results are found in animals treated with chronic neuroleptics (dopamine blockers). The accumulation of dopamine metabolites (dihydroxyphenylalanine and homovanillic acid) in the striatum of diabetic rats is decreased after 4 - 6 wk of diabetes (163). Hyperglycemia suppresses dopamine neuronal firing (164). Furthermore, behavioral responses of diabetic rats to drugs acting through dopamine neuronal systems are increased, consistent with a hypersensitivity seen after decreased dopaminergic neurotransmission (165). Regional noradrenergic transmission is altered in diabetes. Trulson and Himmel (166) showed that forebrain norepinephrine content increases with diabetes and decreases after insulin administration. The major metabolite of norepinephrine (3-methoxy-4-hydroxyphenylglycol sulfate) decreases in diabetes and increases after insulin administration, suggesting that diabetes decreases, whereas hypoglycemia increases forebrain noradrenergic neurotransmission. Steger and Kienast (167) found reduced noradrenergic neurotransmission in STZ-induced diabetes to be at least partly correct with insulin therapy (and may relate to hypogonadism). Chu et al. (168), in regional analyses of brain catecholamines and serotonin levels, found lowered catecholamine levels in the hypothalamus but higher levels in the corpus striatum. They (168) also found lower serotonin levels in both hypothalamus and brain stem but not in the corpus striatum. Reversal of these abnormalities occurs with insulin therapy. These authors also noted several neurological or neurobehavioral changes: STZ-induced diabetic rats had thermoregulatory deficits in the cold, they had a higher spontaneous pain threshold but reduced sensitivity to morphine analgesia, and they had decreased spontaneous motor activity but increased motor responses to amphetamines. Garris (169) found that the norepinephrine content rises in several brain regions in db/db mice compared with age—matched controls. Both oc-1 and -2 receptors are elevated, whereas (3-adrenergic receptors are decreased in regional brain samples of diabetic mice relative to controls. Serotonin neurotransmission is abnormal in experimental diabetes. Crandall and Femstrom (170) found that experimental diabetes alters tryptophan content in the brain (the rate-limiting step in serotonin synthesis is thus affected) because of increased competition by other

564

large neutral amino acids for entry into the brain. MacKenzie and Trulson (171, 172) suggested that reduced serotonin neurotransmission occurs in STZ-induced diabetic rat brain. King and Rohrbach (173) found reduced serotonin synthesis in the hypothalamus of diabetic mice and related this to reduced gonadotropin levels and infertility. In studies of patients recovering from ketoacidosis, Curzon et al. (174) measured cerebrospinal fluid (CSF) levels of two monoamine precursors, tryptophan (precursor to serotonin), and tyrosine (precursor to brain catecholamines including dopamine, norepinephrine, and epinephrine). CSF tryptophan levels increase in diabetic ketoacidosis as do levels of 5-hydroxy-indoleacetic acid, an indicator of serotonin neurotransmission. In addition to these reports of abnormal monoamine neurotransmission in experimental and occasionally human diabetes, there are various reports of abnormalities in several neuropeptides, including neuropeptide Y, substance P, met-enkephalin, somatostatin, vasoactive intestinal peptide (VIP), p-endorphin, and vasopressin (175-179). These neurotransmitters may affect behavior, mood, appetite, and pain perception. What consequences for clinical care of patients with diabetes emanate from these studies? Altered sensitivity to CNS drugs is one possibility. Ganzini et al. (180) suggested that there is an increased risk of tardive dyskinesia in diabetic patients and an increase in movement disorders in diabetic patients after treatment with metoclopramide, a dopamine receptor blocker used to treat gastroparesis diabeticorum. Perhaps, similar altered noradrenergic or serotoninergic sensitivity will also be found in people with long-standing or poorly controlled diabetes. More neuropharmacological studies in people with diabetes seem warranted given the results in animals. DIABETES AND SEIZURES Several early (181-184) and some recent (185-187) reports suggest EEG abnormalities are more common in people with diabetes. The import for clinical medicine of these findings is unclear. Is there an association between diabetes and seizure disorders other than during glycemic extremes? Some antiseizure medications such as phenytoin are potentially diabetogenic (188). Such an association could account for an apparent relationship between diabetes and seizures. The Diabetes Control and Complications Trial provides evidence that tight glucose control increases the risk of seizures (189,190). Undoubtedly, acute hypoglycemia can produce seizures; it also may produce lasting brain damage. As with insulin overtreatment, poorly controlled diabetes may be clinically manifest by seizures in both ketoacidosis (111) and in hyperosmolar coma (191). Do seizure manifestations caused by uncontrolled glycemia predict any chronic increased risk of seizure disorders? The literature is unclear. We hope this issue will be studied in a more systematic fashion.

DIABETES, VOL. 41, MAY 1992

A.L McCALL

A CHRONIC DIABETIC ENCEPHALOPATHY Reske-Nielsen (137) described pathological changes in the nervous system of juvenile IDDM patients of long duration. Quantitative analyses of cortical structure in rats with diabetes for 1 yr found decreased brain volume and weight and loss of cortical neurons (192). Many studies in experimental diabetes found structural and biochemical abnormalities as well (175,176,193-198). As reviewed by Mooradian (1), there are many potential causes for brain dysfunction in diabetes mellitus. These include vascular (stroke, altered blood-brain barrier function) and metabolic (altered glycemia, ketosis, hypoxia, electrolyte and neurotransmitter changes) disturbances and the coexistence of other disorders, e.g., renal failure or hypothyroidism. Several studies found evidence of decreased cognitive performance in patients with diabetes mellitus (2). Although global measures such as intelligence (IQ) tests are often normal, more subtle indices of mental efficiency are frequently disturbed in diabetes. The developments in this research area will, we hope, be correlated with the physiological observations made in humans and animals. The possible consequences to human brain function may thus be better understood both descriptively and mechanistically. By doing so, the potential to prevent brain dysfunction in people with diabetes is enhanced. As reviewed by Ryan (2), IDDM patients may exhibit cognitive deficits. Early onset of diabetes may be a particular risk factor for cognitive impairment. IQ and school achievement tests may be abnormal, particularly if school attendance problems occur. For adults with IDDM, as in NIDDM patients, poor metabolic control predicts decreased cognitive performance. In all groups, recurrent and severe hypoglycemia portends poor mental efficiency. As Ryan points out, much more systematic studies need to be done to sort out the several possible causes of chronic brain damage or dysfunction in diabetes. NIDDM patients have reduced performance on measures of verbal learning, memory retrieval, abstract reasoning, and complex psychomotor performance (2, 199-208). However, not all studies found significant differences in performance for diabetic individuals. Explanations for this discrepancy include the complexity and sophistication of the assessment and the types of patients studied. In general, rather subtle dysfunction is noted in those with higher achievement, not likely to affect daily living skills adversely. In other less functional diabetic patients, more pronounced abnormalities are noted. The specific mechanisms involved in decreased cognitive performance are unclear. Many studies found that poor performance is predicted by poor glycemic control. Higher blood glucose levels and elevated HbAi values result in poorer cognitive test efficiency. Hypertriglyceridemia and the presence of peripheral neuropathy also correlate with poor performance in some studies. If poor diabetic control predicts decreased mental efficiency, how does hyperglycemia affect brain performance? What biochemical mechanisms are responsible? Brain tissues may be subject to nonenzymatic glycosyl-

DIABETES, VOL. 41, MAY 1992

ation (209). The brain seems to be less affected by polyol metabolic abnormalities via the aldose reductase pathway than other tissues (210,211). It is possible that selective CNS cells are impaired, however (212). It would be very helpful if adequate animal models of CNS behavioral dysfunction permitted a more mechanistic determination of involved neurochemical pathways (213). CONCLUSIONS The brain is not spared by diabetes. Clinical complications are manifest in excessive stroke damage, permanent impairment in brain function from hypoglycemia, and a mild chronic encephalopathy. Repeated hypoglycemia as a result of treatment may also interfere with normal protective counterregulatory mechanisms at least partly through CNS effects. It also seems likely that altered responses to neuropharmacological agents and an increased prevalence of seizure disorders may be chronic effects of diabetes on the brain. We understand poorly the basis for these CNS complications of diabetes. Many physiological abnormalities occur in the brain as a chronic consequence of diabetes. CBF is probably decreased by poor diabetes control; cerebrovascular reactivity is clearly impaired in long-standing diabetes mellitus. Neurotransmitter metabolism, action, and binding within the brain are altered at least for monoamines and possibly for other transmitters including neuropeptides. Altered energy metabolism occurs in poorly controlled diabetes. The brain appears to adapt in transport efficiency for metabolic substrates, most notably glucose and ketone bodies, in states of both chronic hyper- and hypoglycemia. This adaptation may partly protect the brain, but it is unlikely that protection is complete. Can we relate the clinical syndromes of CNS complications of diabetes to the physiological changes described? No definitive answers are available. Nonetheless, the evidence suggests that the impact of diabetes on the brain is substantial, although more subtle than some other chronic diabetic complications. Both clinical and basic research needs to be focused on the mechanisms by which abnormal physiology of the brain in diabetes occurs and the best ways to prevent chronic brain damage in patients. Substantial prospects exist to prevent or ameliorate brain damage as a result of diabetes in the future. ACKNOWLEDGMENTS I acknowledge the support of the Juvenile Diabetes Foundation International (JDFI), the joint support of National Institute of Neurological and Communicative Disorders and Stroke, National Institute of Diabetes and Digestive and Kidney Diseases, National Heart, Lung and Blood Institute, and National Institute of Child Health and Human Development (R13-NS-29022), U.S. Public Health Service Grants RO1-NS-22213 and PO1-NS17493, and many colleagues for participation in an International Diabetes Federation Congress Satellite Symposium (29 June to 1 July 1991) on The Central Nervous System and Diabetes from which many ideas for this discussion emanated.

565

DIABETES AND CNS

Particular thanks are expressed to Sara King of the JDFI, Dr. Philip Cryer (Washington Univ., St. Louis, MO), Dr. Robert Sherwin (Yale Univ., New Haven, CT), and the organizing committee members for their help with this symposium.

25. Kozuka M, Smith ML, Siesjo BK: Preischemic hyperglycemia enhances postischemic depression of cerebral metabolic rate. J Cereb Blood Flow Metab 9:478-90, 1989 26. Nedergaard M, Jakobsen J, Diemer NH: Autoradiographic determination of cerebral glucose content, blood flow, and glucose utilization in focal ischemia of the rat brain: influence of the plasma glucose concentration. J Cereb Blood Flow Metab 8:100-108, 1988 27. Nedergaard M, Diemer NH: Focal ischemia of the rat brain, with REFERENCES special reference to the influence of plasma glucose concentra1. Mooradian AD: Diabetes and the central nervous system. Endocr tion. Acta Neuropathol 73:131-37, 1987 Rev 9:346-56, 1988 28. Gjedde A, Siemkowicz E: Effect of glucose and insulin pretreat2. Ryan CM: Neurobehavioral complications of type I diabetes: ment on cerebral metabolic recovery after ischemia. Trans Am examination of possible risk factors. Diabetes Care 11:86-93, Neurol Assoc 103:45-47, 1978 1988 29. Siemkowicz E: The effect of glucose upon restitution after transient 3. McCall AL: Brain microvascular transport and metabolism: implicerebral ischemia: a summary. Acta Neurol Scand 71:417-27, cations for diabetes. In Hyperglycemia, Diabetes and Vascular 1985 Disease. Ruderman NB, Brownlee M, Williamson J, Eds. London, 30. Siemkowicz E, Hansen AJ, Gjedde A: Hyperglycemic ischemia of UK, Oxford Univ. Press, 1992 rat brain: the effect of post-ischemic insulin on metabolic rate. 4. Plum F: The neurologic complications of diabetes mellitus. In Brain Res 243: 386-90, 1982 Diabetes. Williams RH, Ed. New York, Hoeber, 1960, p. 602-22 31. Siemkowicz E, Hansen AJ: Brain extracellular ion composition and 5. Kuller LH, Dorman JS, Wolf PA: Cerebrovascular disease and EEG activity following 10 minutes ischemia in normo- and hyperdiabetes. In Diabetes in America—Diabetes Data Compiled 1984. glycemic rats. Stroke 12:236-40, 1981 Harris Ml, Hamman RF, Eds. Bethesda, MD, 1985, p. 18-1-18-18 (U.S. Dept. of Health and Human Services) 32. Siemkowicz E, Gjedde A: Post-ischemic coma in rat: effect of 6. Abbott RD, Donahue RP, MacMahon SW, Reed DM, Yano K: different pre-ischemic blood glucose levels on cerebral metaDiabetes and the risk of stroke: the Honolulu Heart Program. JAMA bolic recovery after ischemia. Acta Physiol Scand 110:225-32, 257:949-52, 1987 1980 7. Ruderman NB, Haudenschild CC: Diabetes as an atherogenic 33. Siemkowicz E, Hansen AJ: Clinical restitution following cerebral factor. Prog Cardiovasc Dis 26:373-412, 1984 ischemia in hypo-, normo- and hyperglycemic rats. Acta Neurol 8. Stout RW: Insulin and atheroma: 20-yr perspective. Diabetes Care Scand 58:1-8, 1978 13:631-54, 1990 34. de Courten Myers G, Myers RE, Schoolfield L: Hyperglycemia 9. Gwynne JT, McMillan DE: Advances and pathophysiology of enlarges infarct size in cerebrovascular occlusion in cats. Stroke vascular complications of diabetes. Diabetes Care 14:148-52, 19:623-30, 1988 1991 35. Browning RG, Olson DW, Stueven HA, Mateer JR: 50% Dextrose: 10. Coull BM, Beamer N, de Garmo P, Sexton G, Nordt F, Knox R, antidote or toxin? Ann Emerg Med 19:683-87, 1990 Seaman GV: Chronic blood hyperviscosity in subjects with acute 36. Pulsinelli W, Waldman S, Sigsbee B, Rawlinson D, Scherer P, Plum stroke, transient ischemic attack, and risk factors for stroke. Stroke F: Experimental hyperglycemia and diabetes mellitus worsen 22:162-68, 1991 stroke outcome. Trans Am Neurol Assoc 105:21-24, 1980 11. Pulsinelli WA, Levy DE, Sigsbee B, Scherer P, Plum F: Increased 37. Duverger D, MacKenzie ET: The quantification of cerebral infarcdamage after ischemic stroke in patients with hyperglycemia with tion following focal ischemia in the rat: influence of strain, arterial or without established diabetes mellitus. Am J Med 74:540-44, pressure, blood glucose concentration, and age. J Cereb Blood 1983 Flow Metab 8:449-61, 1988 12. Kushner M, Nencini P, Reivich M, Rango M, Jamieson D, Fazekas 38. Collins RC, Dobkin BH, Choi DW: Selective vulnerability of the F, Zimmerman R, Chawluk J, Alavi A, Alves W: Relation of hyperbrain: new insights into the pathophysiology of stroke. Ann Intern glycemia early in ischemic brain infarction to cerebral anatomy, Med 110: 992-1000, 1989 metabolism, and clinical outcome. Ann A/euro/28:129-35, 1990 39. Giffard RG, Monyer H, Choi DW: Selective vulnerability of cultured 13. Berger L, Hakim AM: The association of hyperglycemia with cortical glia to injury by extracellular acidosis. Brain Res 530:138cerebral edema in stroke. Stroke 17:865-71, 1986 41, 1990 14. Helgason CM: Blood glucose and stroke. Stroke 19:1049-53, 40. Pulsinelli WA, Waldman S, Rawlinson D, Plum F: Moderate hyper1988 glycemia augments ischemic brain damage: a neuropathologic 15. Harris Ml, Hadden WC, Knowler WC, Bennett PH: Prevalence of study in the rat. Neurology 32:1239-46, 1982 diabetes and impaired glucose tolerance and plasma glucose 41. Ginsberg MD, Busto R: Rodent models of cerebral ischemia. levels in U.S. population aged 20-74 yr. Diabetes 36:523-34, Stroke 20:1627-42, 1989 1987 42. Zasslow MA, Pearl RG, Shuer LM, Steinberg GK, Lieberson RE, 16. Riddle MC, Hart J: Hyperglycemia, recognized and unrecognized, Larson CP: Hyperglycemia decreases acute neuronal ischemic as a risk factor for stroke and transient ischemic attacks. Stroke changes after middle cerebral artery occlusion in cats. Stroke 13:356-59, 1982 20:519-23, 1989 17. Gray CS, French JM, Bates D, Cartlidge NE, Venables GS, James 43. Wagner KR, Myers RE: Hyperglycemia preserves brain mitochonOF: Increasing age, diabetes mellitus and recovery from stroke. drial respiration during anoxia. J Neurochem 47:1620-26, 1986 Postgrad Med J 65: 720-24, 1989 44. Choi DW: Methods for antagonizing glutamate neurotoxicity. Cere18. Warner DS, Smith ML, Siesjo BK: Ischemia in normo- and hyperbrovasc Brain Metab Rev 2:105-47, 1990 glycemic rats: effects on brain water and electrolytes. Stroke 45. Koh JY, Goldberg MP, Hartley DM, Choi DW: Non-NMDA receptor18:464-71, 1987 mediated neurotoxicity in cortical culture. J Neurosci 10:693-705, 1990 19. Smith ML, von Hanwehr R, Siesjo BK: Changes in extra- and intracellular pH in the brain during and following ischemia in 46. Henneberry RC: Commentary: the role of neuronal energy in the hyperglycemic and in moderately hypoglycemic rats. J Cereb neurotoxicity of excitatory amino acids. Neurobiol Aging 10:611Blood Flow Metab 6:574-83, 1986 13, 1989 20. Koide T, Wieloch TW, Siesjo BK: Chronic dexamethasone pretreat47. Cox JA, Lysko PG, Henneberry RC: Excitatory amino acid neuroment aggravates ischemic neuronal necrosis. J Cereb Blood Flow toxicity at the /V-methyl-D-aspartate receptor in cultured neurons: Metab 6:395-404, 1986 role of the voltage-dependent magnesium block. Brain Res 499: 21. Rehncrona S, Rosen I, Siesjo BK: Brain lactic acidosis and 267-72, 1989 ischemic cell damage. I. Biochemistry and neurophysiology. J 48. Siesjo BK, Bengtsson F: Calcium fluxes, calcium antagonists, and Cereb Blood Flow Metab 1:297-311, 1981 calcium-related pathology in brain ischemia, hypoglycemia, and 22. Siesjo BK: Mechanisms of ischemic brain damage. Crit Care Med spreading depression: a unifying hypothesis. J Cereb Blood Flow 16:954-63, 1988 Metab 9:127-40, 1989 23. Lundgren J, Cardell M, Wieloch T, Siesjo BK: Preischemic hyper49. Berger L, Hakim AM: Nimodipine prevents hyperglycemia-inglycemia and postischemic alteration of rat brain pyruvate dehyduced cerebral acidosis in middle cerebral artery occluded rats. J drogenase activity. J Cereb Blood Flow Metab 10:536-41, 1990 Cereb Blood Flow Metab 9:58-64, 1989 24. Inamura K, Smith ML, Olsson Y, Siesjo BK: Pathogenesis of 50. Kety SS, Woodford RB, Harmel MH, Freyhan FA, Appel KE, substantia nigra lesions following hyperglycemic ischemia: Schmidt CF: Cerebral blood flow and metabolism in schizophrenia: changes in energy metabolites, cerebral blood flow, and morpholthe effects of barbiturate semi-narcosis, insulin coma, and elecogy of pars reticulata in a rat model of ischemia. J Cereb Blood troshock. Am J Psychiatry 104: 765-70, 1948 Flow Metab 8:375-84, 1988 51. Brierley JB: Brain damage due to hypoglycaemia. In: Hypoglycae-

566

DIABETES, VOL. 41, MAY 1992

A.L. McCALL

52. 53.

54. 55.

56. 57.

58.

59.

60. 61. 62. 63. 64. 65.

66.

67. 68.

69.

70. 71. 72. 73. 74. 75.

76.

mia. Marks V, Rose FC, Eds. Oxford, UK, Blackwell, 1981, p. 488-94 Patrick AW, Campbell IW: Fatal hypoglycaemia in insulin-treated diabetes mellitus: clinical features and neuropathological changes. Diabetic Med 7:349-54, 1990 Brierley JB, Brown AW, Meldrum BS, Riche D: The time course of ischaemic neuronal changes in the primate brain following profound arterial hypotension, air embolism and hypoglycaemia. J Physiol 207:59-60P, 1970 Brierley JB, Brown AW, Meldrum BS: The nature and time course of the neuronal alterations resulting from oligaemia and hypoglycaemia in the brain of Macaca mulatta. Brain Res 25:483-99, 1971 McCandless DW, Abel MS: Hypoglycemia and cerebral energy metabolism. In Cerebral Energy Metabolism and Metabolic Encephalopathy. McCandless DW, Ed. New York, Plenum, 1985, p. 27-41 Auer RN, Siesjo BK: Biological differences between ischemia, hypoglycemia, and epilepsy. Ann A/euro/24:699-707, 1988 Kalimo H, Auer RN, Siesjo BK: The temporal evolution of hypoglycemic brain damage. III. Light and electron microscopic findings in the rat caudoputamen. Acta Neuropathol (Berlin) 67:37-50, 1985 Auer RN, Kalimo H, Olsson Y, Siesjo BK: The temporal evolution of hypoglycemic brain damage. II. Light- and electron-microscopic findings in the hippocampal gyrus and subiculum of the rat. Acta Neuropathol (Berlin) 67:25-36, 1985 Auer RN, Kalimo H, Olsson Y, Siesjo BK: The temporal evolution of hypoglycemic brain damage. I. Light- and electron-microscopic findings in the rat cerebral cortex. Acta Neuropathol (Berlin) 67:13-24, 1985 Auer RN, Olsson Y, Siesjo BK: Hypoglycemic brain injury in the rat: correlation of density of brain damage with the EEG isoelectric time: a quantitative study. Diabetes 33:1090-98, 1984 Auer RN, Wieloch T, Olsson Y, Siesjo BK: The distribution of hypoglycemic brain damage. Acta Neuropathol (Berlin) 64:17791, 1984 Auer RN: Progress review: hypoglycemic brain damage. Stroke 17: 699-708, 1986 Pelligrino D, Siesjo BK: Regulation of extra- and intracellular pH in the brain in severe hypoglycemia. J Cereb Blood Flow Metab 1:85-96, 1981 Pelligrino D, Almquist LO, Siesjo BK: Effects of insulin-induced hypoglycemia on intracellular pH and impedance in the cerebral cortex of the rat. Brain Res 221:129-47, 1981 Wieloch T, Harris RJ, Symon L, Siesjo BK: Influence of severe hypoglycemia on brain extracellular calcium and potassium activities, energy, and phospholipid metabolism. J Neurochem 43:16068, 1984 Harris RJ, Wieloch T, Symon L, Siesjo BK: Cerebral extracellular calcium activity in severe hypoglycemia: relation to extracellular potassium and energy state. J Cereb Blood Flow Metab 4:187-93, 1984 Kiessling M, Auer RN, Kleihues P, Siesjo BK: Cerebral protein synthesis during long-term recovery from severe hypoglycemia. J Cereb Blood Flow Metab 6:42-51, 1986 Werner H, Raizada MK, Mudd LM, Foyt HL, Simpson IA, Roberts CT Jr, LeRoith D: Regulation of rat brain/HepG2 glucose transporter gene expression by insulin and insulin-like growth factor-l in primary cultures of neuronal and glial cells. Endocrinology 125: 314-20, 1989 Agardh CD, Kalimo H, Olsson Y, Siesjo BK: Hypoglycemic brain injury: metabolic and structural findings in rat cerebellar cortex during profound insulin-induced hypoglycemia and in the recovery period following glucose administration. J Cereb Blood Flow Metab 1:71-84, 1981 Chapman AG, Westerberg E, Siesjo BK: The metabolism of purine and pyrimidine nucleotides in rat cortex during insulin-induced hypoglycemia and recovery. J Neurochem 36:179-89, 1981 Wieloch T: Hypoglycemia-induced neuronal damage prevented by an A/-methyl-D-aspartate antagonist. Science 230:681-83, 1985 Papagapiou MP, Auer RN: Regional neuroprotective effects of the NMDA receptor antagonist MK-801 (dizocilpine) in hypoglycemic brain damage. J Cereb Blood Flow Metab 10:270-76, 1990 Siesjo BK: Historical overview: Calcium, ischemia, and death of brain cells. Ann NYAcad Sci 522:638-61, 1988 Siesjo BK, Deshpande JK: Electrolyte shifts between brain and plasma in hypoglycemic coma. J Cereb Blood Flow Metab 7:78993, 1987 Langan SJ, Deary IJ, Hepburn DA, Frier BM: Cumulative cognitive impairment following recurrent severe hypoglycaemia in adult patients with insulin-treated diabetes mellitus. Diabetologia 34: 337_44, 1991 Wredling R, Levander S, Adamson U, Lins PE: Permanent neuro-

DIABETES, VOL. 41, MAY 1992

77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94.

95. 96. 97. 98. 99.

100.

101. 102. 103.

104. 105.

psychological impairment after recurrent episodes of severe hypoglycaemia in man. Diabetologia 33:152-57, 1990 Pelligrino DA, Becker GL, Miletich DJ, Albrecht RF: Cerebral mitochondrial respiration in diabetic and chronically hypoglycemic rats. Brain Res 479:241-46, 1989 Pelligrino DA, Segil LJ, Albrecht RF: Brain glucose utilization and transport and cortical function in chronic vs. acute hypoglycemia. Am J Physiol 259:E729-35, 1990 Cryer PE: Decreased sympathochromaffin activity in IDDM. Diabetes 38:405- 409, 1989 Gjedde A, Crone C: Blood-brain glucose transfer: repression in chronic hyperglycemia. Science 214:456-57, 1981 McCall AL, Millington W, Temple S, Wurtman RJ: Altered transport of hexoses across the blood-brain barrier in diabetes (Abstract). Diabetes 28: 381,1979 McCall AL, Millington WR, Wurtman RJ: Metabolic fuel and amino acid transport into the brain in experimental diabetes mellitus. Proc NatlAcad Sci USA 79:5406-10, 1982 Pardridge WM, Triguero D, Farrel CR: Downregulation of bloodbrain barrier glucose transporter in experimental diabetes. Diabetes 39:1040 -44, 1990 Choi TB, Boado RJ, Pardridge WM: Blood-brain barrier glucose transporter mRNA is increased in experimental diabetes mellitus. Biochem Biophys Res Commun 164:375-80, 1989 Mooradian AD, Morin AM: Brain uptake of glucose in diabetes mellitus: the role of glucose transporters. Am J Med Sci 301:17377, 1991 Ruderman NB, Ross PS, Berger M, Goodman MN: Regulation of glucose and ketone-body metabolism in brain of anaesthetized rats. Biochem J 138:1-10, 1974 Gjedde A, Crone C: Blood-brain glucose transfer: repression in chronic hyperglycemia. Science 214:456-57, 1981 Takasato Y, Rapoport SI, Smith QR: An in situ brain perfusion technique to study cerebrovascular transport in the rat. Am J Physiol 247: H484-93, 1984 Harik SI, LaManna JC: Vascular perfusion and blood-brain glucose transport in acute and chronic hyperglycemia. J Neurochem 51:1924-29, 1988 LaManna JC, Harik SI: Regional comparisons of brain glucose influx. Brain Res 326:299-305, 1985 LaManna JC, Harik SI: Regional studies of blood-brain barrier transport of glucose and leucine in awake and anesthetized rats. J Cereb Blood Flow Metab 6:717-23, 1986 Duckrow RB: Glucose transfer into rat brain during acute and chronic hyperglycemia. Metab Brain Dis 3:201-209, 1988 Duckrow RB, Bryan RM Jr: Regional cerebral glucose utilization during hyperglycemia. J Neurochem 48:989-93, 1987 Pelligrino DA, Lipa MD, Albrecht RF: Regional blood-brain glucose transfer and glucose utilization in chronically hyperglycemic, diabetic rats following acute glycemic normalization. J Cereb Blood Flow Metab 10:774-80, 1990 Pardridge WM, Triguero D, Farrell CR: Downregulation of bloodbrain barrier glucose transporter in experimental diabetes. Diabetes 39:1040-44, 1990 McCall AL, Fixman LB, Fleming N, Tornheim K, Chick W, Ruderman NB: Chronic hypoglycemia increases brain glucose transport. Am J Physiol 251: E442-47, 1986 Bryan RM Jr, Pelligrino DA: Cerebral blood flow during chronic hypoglycemia in the rat. Brain Res 475:397-400, 1988 Hara M, Matsuda Y, Okumura N, Hirai K, Nakagawa H: Effect of glucose starvation on glucose transport in neuronal cells in primary culture from rat brain. J Neurochem 52:909-12, 1989 Walker PS, Donovan JA, Van Ness BG, Fellows RE, Pessin JE: Glucose-dependent regulation of glucose transport activity, protein, and mRNA in primary cultures of rat brain glial cells. J Biol Chem 263:15594-601, 1988 Kalckar HM, Ullrey DB: Further clues concerning the vectors essential to regulation of hexose transport, as studied in fibroblast cultures from a metabolic mutant. Proc Natl Acad Sci USA 81: 1126-29, 1984 Haspel HC, Wilk EW, Birnbaum MJ, Cushman SW, Rosen OM: Glucose deprivation and hexose transporter polypeptides of murine fibroblasts. J Biol Chem 261:6778-89, 1986 Mooradian AD: Effect of ascorbate and dehydroascorbate on tissue uptake of glucose. Diabetes 36:1001-1004, 1987 Wurtman RJ, Blusztajn JK, Ulus IH, Coviella IL, Buyukuysal RL, Growdon JH, Slack BE: Choline metabolism in cholinergic neurons: implications for the pathogenesis of neurodegenerative diseases. Adv Neurol 51:117-25, 1990 Knudsen GM, Jakobsen J, Juhler M, Paulson OB: Decreased blood-brain barrier permeability to sodium in early experimental diabetes. Diabetes 35:1371-73, 1986 Knudsen GM, Jakobsen J: Blood-brain barrier permeability to

567

DIABETES AND CNS

106.

107. 108. 109.

110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120.

121.

122.

123.

124. 125. 126.

127. 128.

129.

568

sodium: modification by glucose or insulin. J Neurochem 52:17478,1989 Knudsen GM, Jakobsen J, Barry Dl, Compton AM, Tomlinson DR: Myo-inositol normalizes decreased sodium permeability of the blood-brain barrier in streptozotocin diabetes. Neuroscience 29: 773-77, 1989 Jakobsen J, Knudsen GM, Juhler M: Cation permeability of the blood-brain barrier in streptozotocin-diabetic rats. Diabetologia 30:409-13, 1987 Stauber WT, Ong S-H, McCuskey RS: Selective extravascular escape of albumin into the cerebral cortex of the diabetic rat. Diabetes 30:500-503, 1981 Williamson JR, Chang K, Tilton RG, Prater C, Jeffrey JR, Weigel C, Sherman WR, Eades DM, Kilo C: Increased vascular permeability in spontaneously diabetic BB/W rats and in rats with mild versus severe streptozocin-induced diabetes. Prevention by aldose reductase inhibitors and castration. Diabetes 36:813-21, 1987 McAloon J, Carson D, Crean P: Cerebral oedema complicating diabetic ketoacidosis. Acta Paediatr Scand 79:115-17, 1990 Rosenbloom AL: Intracerebral crises during treatment of diabetic ketoacidosis. Diabetes Car/" 13:22-33, 1990 Van der Meulen JA, Klip A, Grinstein S: Possible mechanism for cerebral oedema in diabetic ketoacidosis. Lancet 2:306-308, 1987 Krane EJ, Rockoff MA, Wallman JK, Wolfsdorf Jl: Subclinical brain swelling in children during treatment of diabetic ketoacidosis. N EnglJ Med 312:1147-51, 1985 Taubin H, Matz R: Cerebral edema, diabetes insipidus, and sudden death during the treatment of diabetic ketoacidosis. Diabetes 17:108 -109, 1968 Duckrow RB, Beard DC, Brennan RW: Regional cerebral blood flow decreases during chronic and acute hyperglycemia. Stroke 18:52-58, 1987 Pelligrino DA, Albrecht RF: Chronic hyperglycemic diabetes in the rat is associated with a selective impairment of cerebral vasodilatory responses. J Cereb Blood Flow Metab 11:667-77, 1991 Jakobsen J, Nedergaard M, Aarslew-Jensen M, Diemer NH: Regional brain glucose metabolism and blood flow in streptozocininduced diabetic rats. Diabetes 39:437-40, 1990 Duckrow RB, Beard DC, Brennan RW: Regional cerebral blood flow decreases during hyperglycemia. Ann Neurol 17:267-72, 1985 Kikano GE, LaManna JC, Harik SI: Brain perfusion in acute and chronic hyperglycemia in rats. Stroke 20:1027-31, 1989 Simpson RE,III, Phillis JW, Buchannan J: A comparison of cerebral blood flow during basal, hypotensive, hypoxic and hypercapnic conditions between normal and streptozotocin diabetic rats. Brain Res 531:136-42, 1990 Wakisaka M, Nagamachi S, Inoue K, Morotomi Y, Nunoi K, Fujishima M: Reduced regional cerebral blood flow in aged noninsulindependent diabetic patients with no history of cerebrovascular disease: evaluation by /V-isopropyl-123l-p-iodoamphetamine with single-photon emission computed tomography. J Diabetic Complications 4:170 -74, 1990 Grill V, Gutniak M, Bjorkman O, Lindqvist M, Stone-Elander S, Seitz RJ, Blomqvist G, Reichard P, Widen L: Cerebral blood flow and substrate utilization in insulin-treated diabetic subjects. Am J Physiol 258:E813 -20, 1990 Neil HA, Gale EA, Hamilton SJ, Lopez Espinoza I, Kaura R, McCarthy ST: Cerebral blood flow increases during insulin-induced hypoglycaemia in type 1 (insulin-dependent) diabetic patients and control subjects. Diabetologia 30:305-309, 1987 Kastrup J, Petersen P, Dejgard A: Intravenous lidocaine and cerebral blood flow: impaired microvascular reactivity in diabetic patients. J Clin Pharmacol 30:318-23, 1990 Griffith DN, Saimbi S, Lewis C, Tolfree S, Betteridge DJ: Abnormal cerebrovascular carbon dioxide reactivity in people with diabetes. Diabetic Med 4:217-20, 1987 Kastrup J, Rrsgaard S, Parving HH, Lassen NA: Impaired autoregulation of cerebral blood flow in long-term type I (insulin-dependent) diabetic patients with nephropathy and retinopathy. Clin Physiol 6:549-59, 1986 Lass P, Knudsen GM, Pedersen EV, Barry Dl: Impaired betaadrenergic mediated cerebral blood flow response in streptozotocin diabetic rats. Pharmacol Toxicol 65:318-20, 1989 Simpson RE III, Phillis JW, Buchannan J: A comparison of cerebral blood flow during basal, hypotensive, hypoxic and hypercapnic conditions between normal and streptozotocin diabetic rats. Brain Res 531:136-42, 1990 Croughwell N, Lyth M, Quill TJ, Newman M, Greeley WJ, Smith LR, Reves JG: Diabetic patients have abnormal cerebral autoregulation during cardiopulmonary bypass. Circulation 82:IV407-12, 1990

130. Paulson OB, Strandgaard S, Edvinsson L: Cerebral autoregulation. Cerebrovasc Brain Metab Rev 2:161-92, 1990 131. Cohen RA, Tesfamariam B, Weisbrod RM, Zitnay KM: Adrenergic denervation in rabbits with diabetes mellitus. Am J Physiol 259: H55-61, 1990 132. Tesfamariam B, Brown ML, Deykin D, Cohen RA: Elevated glucose promotes generation of endothelium-derived vasoconstrictor prostanoids in rabbit aorta. J Clin Invest 85:929-32, 1990 133. Tesfamariam B, Jakubowski JA, Cohen RA: Contraction of diabetic rabbit aorta caused by endothelium-derived PGH2-TxA2. Am J Physiol 257: H1327-31, 1989 134. Jeremy JY, Mikhailidis DP, Thompson CS, Dandona P: Changes in eicosanoid synthesis in the cerebrum, cerebellum and brain stem of the diabetic rat. Diabetes Res 6:95-98, 1987 135. Wey HE, Jakubowski JA, Deykin D: Effect of streptozotocininduced diabetes on prostaglandin production by rat cerebral microvessels. Thromb Res 42:527-38, 1986 136. Mukai N, Hori S, Pomeroy M: Cerebral lesions in rats with streptozotocin-induced diabetes. Acta Neuropathol (Berlin) 51:79-84, 1980 137. Reske-Nielsen E, Lundback K, Rafaelsen OJ: Pathological changes in the central and peripheral nervous system of young long-term diabetics. Diabetologia 1:233-37, 1965 138. Johnson PC, Brendel K, Meezan E: Thickened cerebral cortical capillary basement membranes in diabetics. Arch Pathol Lab Med 106:214-17, 1982 139. McCall AL, Sussman I, Tornheim K, Cordero R, Ruderman NB: Effects of hypoglycemia and diabetes on fuel metabolism by rat brain microvessels. Am J Physiol 25A:E272-78, 1988 140. McCall AL, Gould JB, Ruderman NB: Diabetes-induced alterations of glucose metabolism in rat cerebral microvessels. Am J Physiol 247:E462-67, 1984 141. Hingorani V, Brecher P: Glucose and fatty acid metabolism in normal and diabetic rabbit cerebral microvessels. Am J Physiol 252:E648-53, 1987 142. Kalckar HM: Regulation of hexose transport-carrier activity in cultured animal fibroblasts: another confrontation with cellular recycling requiring oxidative energy generation. Trans NY Acad Sci 41:83-86, 1983 143. Kalckar HM, Ullrey DB: Hexose uptake regulation mediated through aerobic pathways: schism in a fibroblast mutant. Fed Proc 43:2242-45, 1984 144. Ullrey DB, Kalckar HM: Schism and complementation of hexosemediated transport regulation as illustrated in a fibroblast mutant lacking phosphoglucose-isomerase. Biochem Biophys Res Commun 107:1532-38, 1982 145. Ullrey DB, Kalckar HM: The nature of regulation of hexose transport in cultured mammalian fibroblasts: aerobic "repressive" control by D-glucosamine. Arch Biochem Biophys 209:168-74, 1981 146. Kalckar HM, Ullrey DB, Laursen RA: Effects of combined glutamine and serum deprivation on glucose control of hexose transport in mammalian fibroblast cultures. Proc Natl Acad Sci USA 77:595861,1980 147. Kalckar HM, Christopher CW, Ullrey D: Uncouplers of oxidative phosphorylation promote derepression of the hexose transport system in cultures of hamster cells. Proc Natl Acad Sci USA 76:6453-55, 1979 148. Haspel HC, Birnbaum MJ, Wilk EW, Rosen OM: Biosynthetic precursors and in vitro translation products of the glucose transporter of human hepatocarcinoma cells, human fibroblasts, and murine preadipocytes. J Biol Chem 260:7219-25, 1985 149. Kety SS, Polis BD, Nadler CS, Schmidt CF: Blood flow and oxygen consumption of the human brain in diabetic acidosis and coma. J Clin Invest 27:500-10, 1948 150. Kety SS: Circulation and metabolism of the human brain in health and disease. Am J Med 8:205-17, 1950 151. Bradbury MW: The structure and function of the blood-brain barrier. Fed Proc 43:186-90, 1984 152. Blackshear PJ, Alberti KG: Experimental diabetic ketoacidosis: sequential changes of metabolic intermediates in blood, liver, cerebrospinal fluid and brain after acute insulin deprivation in the streptozotocin-diabetic rat. Biochem J 138:107-17, 1974 153. Randle PJ, Garland PB, Hales CN, Newsholme EA: The glucosefatty acid cycle: its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1:785-89, 1963 154. Mooradian AD: Diabetic complications of the central nervous system. Endocr Rev 9:346-56, 1988 155. Mans AM, DeJoseph MR, Davis DW, Hawkins RA: Brain energy metabolism in streptozotocin-diabetes. Biochem J 249:57-62, 1988 156. Krukoff TL, Patel KP: Alterations in brain hexokinase activity associated with streptozotocin-induced diabetes mellitus in the rat. Brain Res 522:157-60, 1990

DIABETES, VOL. 41, MAY 1992

A.L. McCALL

157. Owen OE, Morgan AP, Kemp HG, Sullivan JM, Herrera MG, Cahill GF Jr: Brain metabolism during fasting. J Clin Invest 46:1589-95, 1967 158. Eastman RC, Carson RE, Gordon MR, Berg GW, Lillioja S, Larson SM, Roth J: Brain glucose metabolism in noninsulin-dependent diabetes mellitus: a study in Pima Indians using positron emission tomography during hyperinsulinemia with euglycemic glucose clamp. J Clin Endocrinol Metab 71:1602-10, 1990 159. Grill V: A comparison of brain glucose metabolism in diabetes as measured by positron emission tomography or by arteriovenous techniques. Ann Med 22:171 -76, 1990 160. Gutniak M, Blomqvist G, Widen L, Stone Elander S, Hamberger B, Grill V: D-[U-11C]glucose uptake and metabolism in the brain of insulin-dependent diabetic subjects. Am J Physiol 258:E805-12, 1990 161. Lozovsky D, Sailer CF, Kopin IJ: Dopamine receptor binding is increased in diabetic rats. Science 214:1031-33, 1981 162. Bellush LL, Reid SG: Altered behavior and neurochemistry during short-term insulin withdrawal in streptozocin-induced diabetic rats. Diabetes 40:217'-22, 1991 163. Trulson ME, Himmel CD: Decreased brain dopamine synthesis rate and increased spiroperidol binding in streptozotocin-diabetic rats. J Neurochem 40:1456-59, 1983 164. Sailer CF, Chiodo LA: Glucose suppresses basal firing and haloperidol-induced increases in the firing rate of central dopaminergic neurons. Science 210:1269-71, 1980 165. Sailer CF, Kopin IJ: Glucose potentiates haloperidol-induced catalepsy. LifeSci 29:2337-41, 1981 166. Trulson ME, Himmel CD: Effects of insulin and streptozotocininduced diabetes on brain norepinephrine metabolism in rats. J Neurochem 44: 1873-76, 1985 167. Steger RW, Kienast SG: Effect of continuous versus delayed insulin replacement on sex behavior and neuroendocrine function in diabetic male rats. Diabetes 39:942-48, 1990 168. Chu PC, Lin MT, Shian LR, Leu SY: Alterations in physiologic functions and in brain monoamine content in streptozocin-diabetic rats. Diabetes 35:481-85, 1986 169. Garris DR: Age- and diabetes-associated alterations in regional brain norepinephrine concentrations and adrenergic receptor populations in C57BL/KsJ mice. Brain Res Dev Brain Res 51:161 -66, 1990 170. Crandall E A, Fernstrom JD: Effect of experimental diabetes on the levels of aromatic and branched-chain amino acids in rat blood and brain. Diabetes 32:222-30, 1983 171. MacKenzie RG, Trulson ME: Effects of insulin and streptozotocininduced diabetes on brain tryptophan and serotonin metabolism in rats. J Neurochem 30:205-11, 1978 172. Trulson ME, Jacoby JH, MacKenzie RG; Streptozotocin-induced diabetes reduces brain serotonin synthesis in rats. J Neurochem 46:1068-72, 1986 173. King TS, Rohrbach DH: Reduced aminergic synthesis in the hypothalamus of the infertile, genetically diabetic (C57BL/KsJ-db/ db) male mouse. Exp Brain Res 81:619-25, 1990 174. Curzon G, Kantamaneni BD, Callaghan N, Sullivan PA: Brain transmitter precursors and metabolites in diabetic ketoacidosis. J Neurol Neurosurg Psychiatry 45:489-93, 1982 175. Abbracchio MP, Di Luca M, Di Giulio AM, Cattabeni F, Tenconi B, Gorio A: Denervation and hyperinnervation in the nervous system of diabetic animals. III. Functional alterations of G proteins in diabetic encephalopathy. J Neurosci Res 24:517-23, 1989 176. Di Giulio AM, Tenconi B, La Croix R, Mantegazza P, Abbracchio MP, Cattabeni F, Gorio A: Denervation and hyperinnervation in the nervous system of diabetic animals. II. Monoaminergic and peptidergic alterations in the diabetic encephalopathy. J Neurosci Res 24:362-68, 1989 177. Abe M, Saito M, Ikeda H, Shimazu T: Increased neuropeptide Y content in the arcuato-paraventricular hypothalamic neuronal system in both insulin-dependent and non-insulin-dependent diabetic rats. Brain Res 539:223-27, 1991 178. Timmers Kl, Palkovits M, Coleman DL: Unique alterations of neuropeptide content in median eminence, amygdala, and dorsal vagal complex of 3- and 6-week-old diabetes mutant mice. Metabolism 39:1158-66, 1990 179. Fernstrom JD, Fernstrom MH, Kwok RP: In vivo somatostatin, vasopressin, and oxytocin synthesis in diabetic rat hypothalamus. Am J Physiol 258:E661-66, 1990 180. Ganzini L, Heintz RT, Hoffman WF, Casey DE: The prevalence of tardive dyskinesia in neuroleptic-treated diabetics: a controlled study. Arch Gen Psychiatry 48:259-63, 1991 181. Ellenberg M, Pollack H: Convulsive state in diabetes. Am J Med Sci 214:503, 1947 182. Greenblatt M, Murry J, Root HF: EEG studies in diabetes mellitus. N Engl J Med 234:119, 1946

DIABETES, VOL. 41, MAY 1992

183. Fabrykant M, Pacella BL: Labile diabetes: EEG status and effect of anti-convulsive therapy. Ann Intern Med 29:860, 1948 184. Izzo JL, Schuster DB, Engel GL: The electroencephalogram of patients with diabetes mellitus. Diabetes 2:93, 1953 185. Fisher BM, Frier BM: Nocturnal convulsions and insulin-induced hypoglycaemia in diabetic patients. Postgrad Med J 63:673-76, 1987 186. Malouf R, Brust JC: Hypoglycemia: causes, neurological manifestations, and outcome. Ann Neurol 17:421-30, 1985 187. Husband DJ, Gill GV: "Sunbed seizures": a hypoglycaemic hazard for insulin-dependent diabetics (Letter). Lancet 2:1477, 1984 188. Carter BL, Small RE, Mandel MD, Starkman MT: Phenytoin-induced hyperglycemia. Am J Hosp Pharm 38:1508-12, 1981 189. The DCCT Research Group : Diabetes Control and Complications Trial (DCCT): results of feasibility study. Diabetes Care 10:1-19, 1987 190. The DCCT Research Group: Epidemiology of severe hypoglycemia in the diabetes control and complications trial. Am J Med 90:450-59, 1991 191. Guisado R, Arieff Al: Neurologic manifestations of diabetic comas: correlation with biochemical alterations in the brain. Metabolism 24:665-79, 1975 192. Jakobsen J, Sidenius P, Gundersen HJ, Osterby R: Quantitative changes of cerebral neocortical structure in insulin-treated longterm streptozocin-induced diabetes in rats. Diabetes 36:597-601, 1987 193. Bestetti G, Hofer R, Rossi GL: The preoptic-suprachiasmatic nuclei though morphologically heterogeneous are equally affected by streptozotocin diabetes. Exp Brain Res 66:74-82, 1987 194. Affolter V, Boujon P, Bestetti G, Rossi GL: Hypothalamic and cortical neurons of normotensive and spontaneously hypertensive rats are differently affected by streptozotocin diabetes. Acta Neuropathol 70:135-41, 1986 195. Bestetti G, Locatelli V, Tirone F, Rossi GL, Muller EE: One month of streptozotocin-diabetes induces different neuroendocrine and morphological alterations in the hypothalamo-pituitary axis of male and female rats. Endocrinology 117:208-16, 1985 196. Junker U, Jaggi C, Bestetti G, Rossi GL: Basement membrane of hypothalamus and cortex capillaries from normotensive and spontaneously hypertensive rats with streptozotocin-induced diabetes. Acta Neuropathol 65:202-208, 1985 197. Bestetti G, Rossi GL: Hypothalamic changes in diabetic Chinese hamsters: a semiquantitative, light and electron microscopic study. Lab Invest 47:516-22, 1982 198. Rossi GL, Bestetti G: Morphological changes in the hypothalamichypophyseal-gonadal axis of male rats after twelve months of streptozotocin-induced diabetes. Diabetologia 21:476-81, 1981 199. Franceschi M, Cecchetto R, Minicucci F, Smizne S, Baio G, Canal N: Cognitive processes in insulin-dependent diabetes. Diabetes Care 7:228-31, 1984 200. Tun PA, Perlmuter LC, Russo P, Nathan DM: Memory self-assessment and performance in aged diabetics and non-diabetics. Exp Aging Res 13:151-57, 1987 201. Perlmuter LC, Nathan DM, Goldfinger SH, Russo PA, Yates J, Larkin M: Triglyceride levels affect cognitive function in noninsulindependent diabetics. J Diabetic Complications 2:210-13, 1988 202. Mooradian AD, Perryman K, Fitten J, Kavonian GD, Morley JE: Cortical function in elderly non-insulin dependent diabetic patients: behavioral and electrophysiologic studies. Arch Intern Med 148: 2369-72, 1988 203. Tun PA, Nathan DM, Perlmuter LC: Cognitive and affective disorders in elderly diabetics. Clin Geriatr Med 6:731-46, 1990 204. Reaven GM, Thompson LW, Nahum D, Haskins E: Relationship between hyperglycemia and cognitive function in older NIDDM patients. Diabetes Care 13:16-21, 1990 205. Perlmuter LC: Choice enhances performance in non-insulin dependent diabetics and controls. J Geronto/46:P218-23, 1991 206. Dejgaard A, Gade A, Larsson H, Balle V, Parving A, Parving HH: Evidence for diabetic encephalopathy. Diabetic Med 8:162-67, 1991 207. Holmes CS, Richman LC: Cognitive profiles of children with insulindependent diabetes. J Dev Behav Pediatr 6:323-26, 1985 208. Holmes CS, Koepke KM, Thompson RG, Gyves PW, Weydert JA: Verbal fluency and naming performance in type I diabetes at different blood glucose concentrations. Diabetes Care 7:454-59, 1984 209. Brownlee M, Cerami A, Vlassara H: Advanced glycosylation end products in tissue and the biochemical basis of diabetic complications. N EnglJ Med 318:1315-21, 1988 210. Sredy J, Sawicki DR, Notvest RR: Polyol pathway activity in nervous tissues of diabetic and galactose-fed rats: effect of dietary galactose withdrawal or tolrestat intervention therapy. J Diabetic Complications 5:42-47, 1991

569

DIABETES AND CNS

211. Tilton RG, Chang K, Pugliese G, Eades DM, Province MA, Sherman WR, Kilo C, Williamson JR: Prevention of hemodynamic and vascular albumin filtration changes in diabetic rats by aldose reductase inhibitors. Diabetes 38: 1258-70, 1989 212. Sussman I, Carson MP, Schultz V, Wu XP, McCall AL, Ruderman NB, Tornheim K: Chronic exposure to high glucose decreases myoinositol in cultured cerebral microvascular pericytes but not in endothelium. Diabetologia 31:771-75, 1988 213. Johansson B, Meyerson B, Eriksson UJ: Behavioral effects of an intrauterine or neonatal diabetic environment in the rat. Biol Neonate 59: 226-35, 1991

570

214. Mitrakou A, Ryan C, Veneman T, Mokan M, Jenssen T, Kiss I, Durrant J, Cryer P, Gerich J: Hierarchy of glycemic thresholds for counterregulatory hormone secretion, symptoms, and cerebral dysfunction. Am J Physiol 260:E67-74, 1991 215. Jones TW, McCarthy G, Tamborlane WV, Caprio S, Roessler E, Kraemer D, Starick Zych K, Allison T, Boulware SD, Sherwin RS: Mild hypoglycemia and impairment of brain stem and cortical evoked potentials in healthy subjects. Diabetes 39:1550-55,1990 216. Blackman JD, Towle VL, Lewis GF, Spire JP, Polonsky KS: Hypoglycemic Thresholds for Cognitive Dysfunction in Humans. Diabetes 39:828-35, 1990

DIABETES, VOL 41, MAY 1992

The impact of diabetes on the CNS.

The brain is not usually thought to be a target of chronic diabetes complications. Nonetheless, substantial evidence, summarized herein, suggests that...
2MB Sizes 0 Downloads 0 Views