• Non ci sono risultati.

CHAPTER 3 Plasma

N/A
N/A
Protected

Academic year: 2021

Condividi "CHAPTER 3 Plasma"

Copied!
14
0
0

Testo completo

(1)

CHAPTER 3

Plasma β-amyloid levels in drug-resistant bipolar depressed patients receiving electroconvulsive therapy (Neuropsychobiology 2013; 67(4):185-191)

Authors: Piccinni A, Veltri A, Vizzaccaro C, Catena Dell’Osso M, Medda P, Domenici L, Vanelli F, Cecchini M, Franceschini C, Conversano C, Marazziti D, Dell'Osso L.

ABSTRACT

Aims: Alterations of plasma amyloid-β (Aβ) peptides have been related to a high risk for cognitive impairment and dementia. The present study aimed to measure plasma Aβ peptides (Aβ40, Aβ42) and the Aβ40/Aβ42 ratio in a sample of drug-resistant bipolar depressed patients, as well as to explore the possible correlation between biological parameters and clinical changes along an electroconvulsive therapy (ECT) course. Methods: Aβ40 and Aβ42 were measured by means of an ELISA assay in 25 drug-resistant bipolar depressed patients before (T0) and 1 week after (T1) the end of ECT. The patients were clinically evaluated by means of the Hamilton Rating Scale for Depression, 21-item (HRSD-21), the Mini-Mental State Examination, and the Clinical Global Impressions-Severity of Illness Scale. Results: Plasma Aβ levels and the Aβ40/Aβ42 ratio were similar at T0 and T1. The Aβ40/Aβ42 ratio correlated positively with the HRSD total score at both T0 and T1. At T0, a negative correlation was found between the Aβ40/Aβ42 ratio and the improvement of depressive and cognitive symptoms. Moreover, remitters (n = 9; HRSD ≤ 10) showed a significantly lower Aβ40/Aβ42 ratio at T0 than non-remitters. Conclusion: The present data suggest that a low Aβ40/Aβ42 ratio might characterize a subgroup of depressed patients who respond to ECT, while higher values of this parameter seem to be typical of more severe cases of patients with cognitive impairment.

INTRODUCTION

The risk for dementia and cognitive deterioration is greater in patients affected by mood disorders than in the general population (Geerlings et al., 2008; Gualtieri and Johnson, 2008; Barnes et al., 2012), and it has been related to the number of affective episodes, manic polarity, and the presence of psychotic symptoms (Kessing and Andersen, 2004; Robinson et al., 2006; Torres et al., 2007). The cognitive decline in mood disorders has been interpreted as the result of a common neuropathological mechanism, or as the expression of a greater vulnerability of some affective patients towards neurodegenerative phenomena (Aznar and Knudsen, 2011)

(2)

Amyloid-β (Aβ) deposition in Alzheimer’s disease (AD) is characterized by the presence of fragments of various lengths. In particular, the Aβ42 aminoacid peptide is the main component of senile plaques and is considered the most neurotoxic (Selkoe 2006), whereas the Aβ40 amino acid peptide represents a component of amyloid-associated brain angiopathy (Zhang-Nunes et al., 2006). Indeed, an increase in Aβ40 plasma levels has been associated with microvascular brain pathology, white matter hyperintensities, and lacunar infarcts (van Dijk et al., 2004; Gurol et al., 2006), all conditions related to cognitive decline in the elderly (Longstreth et al., 1996), risk for dementia (Vermeer et al., 2003), and depressive symptoms (de Groot et al., 2000). Most of the studies in AD and mild cognitive impairment reported a reduction of plasma Aβ42 and an increase of Aβ40 and Aβ40/Aβ42 ratio (Van Oijen et al., 2006; Graff-Radford et al., 2007; Xu et al., 2008). However, a few data are available regarding the relationship between depression and peripheral levels of Aβ peptides. Higher levels of Aβ40, lower levels of Aβ42, and a greater Aβ40/Aβ42 ratio have been reported in elderly patients with depression, as compared with healthy control subjects (Qiu et al., 2007; Sun et al., 2007, 2008). Moreover, the study by Sun et al. (2008) highlighted how the so-called ‘amyloid-associated’ depression (with high Aβ40/Aβ42 ratio) is characterized by a more severe deficit in memory, visual-spatial skills, and executive functions. In another study (Kita et al., 2009), depressed patients showed an enhanced Aβ40/Aβ42 ratio, but no differences in Aβ42 levels, when compared with age-matched control subjects. In our recent study on bipolar depressed patients, a negative correlation was found between Aβ42 plasma levels and duration of illness, and a positive one between the Aβ40/Aβ42 ratio and the number of affective episodes (Piccinni et al., 2012).

Since over 70 years, electroconvulsive therapy (ECT) represents one of the most effective treatments for unipolar (Pagnin et al., 2004) and bipolar depression (Zomberg and Pope 1993; Daly et al., 2001) with an effectiveness rate greater than 60%, although its mechanism of action is still unknown. Several factors including sex, age, diagnosis, presence of psychosis, duration of the index episode, medication treatment failure prior to ECT, and medications during the ECT course, have been proposed as predictors of response to ECT in patients suffering from depression (Bloch et al., 2005; Kho et al., 2005; Pluijms et al., 2006), although some disagreements exist (Dombrovski et al., 2005). On the contrary, data on potential biological predictors for the efficacy of ECT are scarce.

Given the paucity of available data, the aim of the present study was to assess plasma Aβ40 and Aβ42 levels and the Aβ40/Aβ42 ratio in bipolar depressed patients who failed to respond

(3)

to medications and received ECT. Possible correlations between biological parameters and clinical changes after the ECT course were examined.

METHODS Subjects

Twenty-five patients (13 men and 12 women; mean age ± SD: 44.1 ± 11.9 years) suffering from bipolar I or II depressive episodes with or without psychotic symptoms according to DSM-IV-TR criteria, were selected at the outpatients’ ward and hospital of the Department of Psychiatry of the University of Pisa. Subjects were eligible if they were over 18 years of age, were not suffering from a major neurological or medical illness that limited the use of ECT, and had no history of substance abuse in the last 6 months. Inclusion criteria were failure to respond to at least three mood disorder treatments, comprising an adequate trial with a tricyclic antidepressant (TCA) (Thase and Rush, 1997; Fava, 2003). More specifically, treatment nonresponsiveness in patients with bipolar depression was defined as persisting depressive symptoms despite two trials of at least 8 weeks, consisting of one trial with mood stabilizer(s) plus a TCA (200 mg/day of imipramine or the equivalent, or the maximum tolerable dose) and one trial with a selective serotonin reuptake inhibitor (40 mg/day of fluoxetine or the equivalent) combined with mood stabilizer(s) and a TCA. Diagnosis was confirmed by means of the Mini International Neuropsychiatric Interview (MINI) (Sheehan et al., 1998). For each patient, a senior psychiatrist recommended ECT according to clinical judgment based on the patient’s failure to respond to medication trials and severity or urgency of illness. A written informed consent was obtained from all subjects, which was approved by the Ethics Committee of the University of Pisa, after procedure and effects were fully explained.

Clinical assessment

Severity of depression was assessed by means of the Hamilton Rating Scale for Depression, 21-item (HRSD-21) (Hamilton, 1960) and the Clinical Global Impressions-Severity of Illness Scale (CGI-S) (Guy, 1976). Remission was considered when HRSD total scores were ≤ 10 at the last two consecutive assessments (Lecrubier, 2002). The sample was screened for significant cognitive impairment by means of the Mini-Mental State Examination (MMSE) (Folstein et al., 1975).

Patients were clinically evaluated by means of HRSD-21, CGI-S, and MMSE before the first ECT session (T0) and 1 week after (T1) the last ECT session. Together with clinical assessment, a blood sample was collected to measure plasma Aβ40 and Aβ42 levels. We

(4)

decided to collect blood at T0 within 30 min before the ECT session to avoid a possible acute effect of electroconvulsion on peripheral Aβ levels. In a previous study, Zimmermann et al. (Zimmermann et al., 2012) hypothesized that the acute variations of peripheral Aβ levels after ECT could depend not only on an increased release from neurons, but also from peripheral sources, like thrombocytes or muscles. This last effect could represent a confounding factor and an important bias.

In order to measure the amount of time the patients spent being ill, the parameter ‘duration of illness’ was assessed according to the following equation: (current age – onset age) × 100/current age, developed by us.

Moreover, two parameters were elaborated by our statistician to assess the changes of depressive and cognitive symptoms: the percentage of depression improvement (PDI) calculated according to the following equation: [(HRSD total score)T0 – (HRSD total score)T1] × 100/(HRSD total score)T0, and the percentage of cognitive improvement (PCI) calculated as: [(MMSE total score) T1 – (MMSE total score)T0] × 100/30-(MMSE total score)T0.

ECT treatment procedures

Before undergoing ECT, each patient was screened for general medical conditions by means of an accurate clinical evaluation including collection of a detailed medical history, a physical and neurological examination, blood and urine tests, electrocardiogram, chest X-ray, and a cerebral computed tomography scan. Anesthesia was induced with intravenous thiopental (2–4 mg/kg i.v.) and succinylcholine (0.5–1 mg/kg i.v.). Bilateral brief-pulse, square-wave stimuli were delivered using a Mecta Spectrum 5000 (Mecta Corporation, Lake Oswego, Oreg., USA) twice a week between 7:00 and 9:00 a.m. Stimulus setting was initially based on age, and the length of the seizures, measured by an electroencephalogram, was kept above 25 s. If the duration of the seizure fell below 25 s, the stimulus was raised (1.5 times) at the next session. For all patients, the electrode positioning and stimulus dosing were similar. Other physiological parameters were monitored by means of a pulse oximetry and an electrocardiogram. Patients were ventilated with 100% oxygen until resumption of spontaneous respiration. ECT treatment was completed on the basis of the clinical judgment of the treating clinicians. The mean number of treatments received was 8.3 (range 6–10). Patients were maintained on the same treatment for at least 4 weeks before and throughout the entire study period. Mood stabilizers were discontinued for 3 days before starting ECT and were withheld until the end of ECT treatment.

(5)

Aβ assay

All study participants were subjected to a blood draw (16-ml test tube) for the measurements of Aβ plasma levels. Samples were centrifuged at 2,500 rpm × 10 min. The plasma was divided into aliquots and preserved at –80 ° C. Analyses of the Aβ40 and Aβ42 levels were carried out using specific ELISA kits (Invitrogen, Camarillo, Calif., USA). Briefly, samples were incubated with the monoclonal antibody specific for the NH2-terminus region of human Aβ that was coated onto the microplate wells. During the first incubation, standard, control, and unknown samples were pipetted into the wells and co-incubated with a rabbit antibody specific for the COOH-terminus of the 1–42 or 1–40 Aβ sequence. Bound rabbit antibody was detected by the use of a horsera dish peroxidase-labeled anti-rabbit antibody. After washing, horseradish peroxidase-labeled anti-rabbit antibody (enzyme) was added. After a second incubation and washing to remove all unbound enzyme, a substrate solution was added, which is acted upon by the bound enzyme to produce color. The intensity of this colored product is directly proportional to the concentration of human Aβ42 or Aβ40 present in the original specimen. Absorption was read at 450 nm using a microplate reader (iMARK; Biorad Laboratories). Samples were quantitated by the standard curve generated from the recombinant Aβ42 or Aβ40 included with the kit.

Statistical analysis

Data were recorded in a specifically designed database and elaborated by means of SPSS software (version No. 17). The comparison between quantitative variables of non-Gaussian distribution was performed with nonparametric statistical tests: in particular, the Mann-Whitney test was used for the comparison of independent samples, while the Wilcoxon test was used to compare the paired data. Correlations between the variables were examined by means of Spearman’s coefficient. The χ 2 test was used to compare categorical variables, while the paired data with Gaussian distribution were compared by paired samples Student’s t-test.

RESULTS

Although all patients showed a clinical improvement, as assessed by the changes in rating scale scores, only 9 patients reached the clinical remission after ECT (HRSD ≤ 10). The HRSD total score (mean values ± SD) significantly decreased from T0 to T1 (T0: 30.5 ± 4.30; T1: 11.4 ± 2.6; t = –30.085, p < 0.0001), and the same was true for the CGI-S score (mean values ± SD) (T0: 5.8 ± 0.4; T1: 2.9 ± 0.8; t = –18.437, p < 0.0001).

(6)

On the contrary, the MMSE total score (mean values ± SD) significantly increased from T0 to T1 (T0: 23.4 ± 2.1; T1: 24.6 ±1.9; t = 4.748, p = 0.0001).

As shown in Table 1 , plasma Aβ40 levels, Aβ42 levels and the Aβ40/Aβ42 ratio were similar at T0 and T1.

Correlation analyses showed that the Aβ40/Aβ42 ratio correlated positively with the HRSD total score at both T0 (rs = 0.562, p = 0.003) and T1 (rs = 0.738, p < 0.001), and negatively with the MMSE total score at T1 only (rs = –0.559, p = 0.004). A significant and positive correlation was also detected between plasma Aβ40 levels and the HRSD total score at both T0 (rs = 0.510, p = 0.009) and T1 (rs = 0.531, p = 0.006).

Finally, a significant and negative correlation was found between plasma Aβ40 levels and the MMSE total score at both T0 (rs = –0.542, p = 0.006) and T1 (rs = –0.548, p = 0.006).

In addition, the sample was characterized by a duration of illness (mean ± SD) of 36.10 ± 17.37% and by a number of episodes (mean ± SD) of 4.12 ± 1.83. Plasma Aβ42 levels were negatively correlated with the duration of illness (rs = − 0.413, p = 0.04), while the Aβ40/Aβ42 ratio was positively correlated with the number of affective episodes (rs = 0.540, p = 0.005). As far as the ECT response was concerned, the Aβ40/Aβ42 ratio at T0 was negatively correlated with PDI (rs = –0.463, p = 0.02) and PCI (rs = –0.637, p = 0.001) (Table 2).

(7)

Moreover, remitters (HRSD ≤ 10, n = 9) at the end of the ECT course, were those with a lower Aβ40/Aβ42 ratio at T0 than the others (z = –2.46, p = 0.014) (Fig. 1).

Figure 1 - Aβ40/Aβ42 ratio (T0) in remitter and non-remitters. At baseline (T0), the group of remitters showed a mean Aβ40/Aβ42 ratio significantly lower than that of non-remitters. * Mann-Whitney test: z = –2.46, p = 0.014.

DISCUSSION

Our study aimed to assess plasma Aβ peptides in a sample of bipolar depressed patients before and after an ECT course. Our findings showed no changes in Aβ42 or Aβ40 levels, as well as in the Aβ40/Aβ42 ratio at the different assessment times. The stability of the Aβ levels, despite the clinical improvement following ECT, could suggest the need for further investigation in

(8)

order to understand if the alterations of peripheral Aβ peptides in patients suffering from bipolar depression might represent a trait rather than a state biomarker. This finding can be considered in line with that of a recent study demonstrating only a slight (between 5 and 10%) and transient (reversible within 2 h) increase of plasma Aβ peptide concentrations after single sessions of ECT. This might be due to a decreased integrity of the blood-brain barrier permeability, an increased release of Aβ from the neurons, or from peripheral sources, like thrombocytes or muscles (Zimmermann et al., 2012). However, to the best of our knowledge, our study is the first evaluating Aβ levels before and after ECT. In the literature, just a few indirect observations are available, like lower Aβ40 levels in patients receiving antidepressants (Sun et al., 2007), or no differences in plasma Aβ42 levels between patients with or without antidepressants (Sun et al., 2007; Pomara et al., 2006). Recently, no influence of antidepressants, lithium, or valproate treatments on the Aβ40/Aβ42 ratio was described in a large sample of depressed patients (Baba et al., 2012).

Our results showed that both Aβ40 levels and the Aβ40/Aβ42 ratio were positively correlated with the severity of depressive and cognitive symptoms, that is to say, the higher the biological parameters, the most severe the patient, and vice versa. According to some authors, the presence of depressive symptoms associated with a high Aβ40/Aβ42 ratio would represent a prodromal manifestation of AD (Sun et al., 2008). However, other hypotheses could be suggested, such as the existence of a primarily ‘amyloid-associated’ mood disorder (characterized by Aβ disturbances unrelated to prodromal AD) with resistance to treatment and risk for cognitive decline (Pomara and Murala Doraiswamy, 2003; Pomara and Sidtis, 2010). Alternatively, Aβ-mediated neurodegenerative processes could be triggered by recurrence of affective episodes representing an epiphenomenon in all patients with mood disorders. Interestingly, in both this study and in another of our group (Piccinni et al., 2012) a positive correlation was detected between the Aβ40/Aβ42 ratio and the number of affective episodes, while highlighting how the changes of blood Aβ levels could characterize a subgroup of bipolar patients with worse illness course.

In the present study, high values of the Aβ40/Aβ42 ratio were associated with a poor outcome of depressive and cognitive symptoms. Moreover, remitters presented a significantly lower Aβ40/Aβ42 ratio at baseline, as compared with non-remitters. Further studies are, however, needed to investigate whether the Aβ-mediated neurotoxicity might contribute to the worsening and subsequent poor response to treatments in affective patients. Although the mode of action of ECT is still elusive, different data show that electroconvulsive seizures may provoke a robust neurotrophic effect by increasing the levels of brain-derived neurotrophic

(9)

factor (BDNF) mRNA, proteins, and the tyrosine kinase receptor B mRNA in the rat hippocampus. Further, chronic electroconvulsive seizure administration blocked the downregulation of BDNF mRNA in the same area in response to restraint-induced stress (Lindefors et al., 1995; Nibuya et al., 1995; Angelucci et al., 2002). Some studies in depressed patients showed that ECT similarly to antidepressants (Piccinni et al., 2008; Sen et al., 2008) may increase the amount of serum and plasma BDNF (Bocchio-Chiavetto et al., 2006; Marano et al., 2007; Taylor, 2008) particularly in responders (Okamoto et al., 2008; Piccinni et al., 2009). These results suggest that one of the putative mechanisms of ECT might be mediated by BDNF and related substances (Taylor, 2008). It is therefore plausible that Aβ may exhibit a functional interference with BDNF actions. In fact, in cortical neuron cultures, subtoxic concentrations of Aβ impair the transduction pathways activated by BDNF signalling (Tong et al., 2001) and, at higher concentrations, the synthesis of BDNF (Arvanitis et al., 2007; Arancio and Chao, 2007). In rats, a BDNF pretreatment is able to prevent neuronal damage induced by Aβ injection (Arancibia et al., 2008), and a single i.v. Aβ injection was reported to induce a depressive phenotype and to inhibit BDNF expression in the prefrontal cortex (Colaianna et al., 2010). Based on these observations, we speculate with caution that the altered plasma Aβ peptide levels of the resistant patients would reflect some brain neurodegenerative and neurotoxic phenomena hindering the BDNF-mediated neurorestorative action induced by ECT. This could explain why in our sample a high Aβ40/Aβ42 ratio seems to be predictive of nonresponse to ECT.

The small sample size is the major bias of the present study. For this reason, the comparisons of biological parameters between remitters and non-remitters are based on a few cases (9 vs. 16 subjects). Further, the fact that all the patients were taking psychotropic drugs during the ECT course may constitute a possible bias, although no influence of psychotropic medications on the plasma Aβ40/Aβ42 ratio was reported (Baba et al., 2012). Moreover, the precise origin of plasma Aβ is not known and results regarding the relationship between peripheral blood and cerebrospinal fluid Aβ levels have been contradictory (Mehta et al., 2001; Kawarabayashi et al., 2001; Giedraitis et al., 2007; Le Bastard et al., 2009). Therefore, a lack of data on cerebrospinal fluid may also be considered a limitation. Lastly, further studies comparing baseline plasma Aβ levels of patients with those of healthy control subjects are needed to clarify whether potential variations of Aβ peptides to ECT could depend on baseline values, and therefore whether or not originally normal or abnormal plasma Aβ peptide levels might influence this response.

(10)

CONCLUSIONS

In conclusion, we propose with caution that particular changes in Aβ peptide levels may represent a potential trait marker of a mood disorder subtype characterized by a great severity, a high cognitive impairment, and worse response to treatments. In any case, studies with larger samples of patients suffering from mood disorders are needed to understand whether changes in plasma Aβ peptides may be used as possible predictors of response, are associated with alterations of other biomarkers, such as the BDNF, and might, therefore, represent a trait or state marker of mood disorders. Similarly, it would be extremely useful to explore more thoroughly the possible associations of Aβ peptides with specific symptoms or symptom clusters, irrespective of the diagnosis.

REFERENCES

Angelucci F, Aloe L, Jiménez-Vasquez P, Mathé AA. Electroconvulsive stimuli alter the regional concentrations of nerve growth factor, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor in adult rat brain. J ECT 2002; 18: 138-143.

Arancibia S, Silhol M, Moulière F et al. Protective effect of BDNF against beta-amyloid induced neurotoxicity in vitro and in vivo in rats. Neurobiol Disease 2008; 31: 316-326.

Arancio O, Chao MV. Neurotrophins, synaptic plasticity and dementia. Curr Op Neurobiol 2007; 17: 325-330.

Arvanitis DN, Ducatenzeiler A, Ou JN et al. High intracellular concentrations of amyloid-beta block nuclear translocation of phosphorylated CREB. J Neurochem 2007; 103(1): 216-228. Aznar S, Knudsen GM. Depression and Alzheimer's disease: is stress the initiating factor in a common neuropathological cascade? J Alzheimers Dis 2011; 23(2): 177-193.

Baba H, Nakano Y, Maeshima H et al. Metabolism of Amyloid-β protein may be affected in depression. J Clin Psichiatry 2012; 73(1): 115-120.

Barnes DE, Yaffe K, Byers AL et al. Midlife vs late-life depressive symptoms and risk of dementia: differential effects for Alzheimer disease and vascular dementia. Arch Gen Psychiatry 2012; 69(5): 493-498.

Bloch Y, Ratzoni G, Sobol D et al. Gender differences in electroconvulsive therapy: a retrospective chart review. J Affect Disord 2005; 84: 99-102.

Bocchio-Chiavetto L, Zanardini R, Bortolomasi M et al. Electroconvulsive therapy (ECT) increases serum brain derived neurotrophic factor (BDNF) in drug resistant depressed patients. Eur Neuropsychopharmacol 2006; 16: 620-624.

(11)

Colaianna M, Tucci P, Zotti M et al. Soluble beta amyloid(1-42): a critical player in producing behavioral and biochemical changes evoking depressive-related state? Br J Pharmacol 2010; 159(8): 1704-1715.

Daly JJ, Prudic J, Devanand DP et al. ECT in bipolar and unipolar depression: differences in speed of response. Bipolar Disord 2001; 3: 95-104.

de Groot JC, de Leeuw FE, Oudkerk M et al. Cerebral white matter lesions and depressive symptoms in elderly adults. Arch Gen Psychiatry 2000; 57: 1071-1076.

Dombrovski AY, Mulsant BH, Haskett RF et al. Predictors of remission after electroconvulsive therapy in unipolar major depression. J Clin Psychiatry 2005; 66: 1043-1049. Fava M. Diagnosis and definition of treatment-resistant depression. Biol Psychiatry 2003; 53: 649-659.

Folstein MF, Folstein SE, McHugh PR. “Mini-Mental State”: a practical method for grading the cognitive state of patients for the clinician. J Psychiat Res 1975; 12: 189-198.

Geerlings MI, den Heijer T, Koudstaal PJ, Hofman A, Breteler MM. History of depression, depressive symptoms, and medial temporal lobe atrophy and the risk of Alzheimer disease. Neurology 2008; 70: 1258-1264.

Giedraitis V, Sundelöf J, Irizarry MC et al. The normal equilibrium between CSF and plasma amyloid beta levels is disrupted in Alzheimer's disease. Neurosci Lett 2007;12;427(3): 127-131.

Graff-Radford NR, Crook JE, Lucas J et al. Association of low plasma Abeta42/Abeta40 ratios with increased imminent risk for mild cognitive impairment and Alzheimer disease. Arch Neurol 2007; 64: 354-362.

Gualtieri CT, Johnson LG. Age-related cognitive decline in patients with mood disorders. Progr Neuropsychopharmacol Biol Psychiatry 2008; 32: 962-967.

Gurol ME, Irizarry MC, Smith EE et al. Plasma beta-amyloid and white mat ter lesions in AD, MCI, and cerebral amyloid angiopathy. Neurology 2006; 66: 23-29.

Guy W. ECDEU assessment manual for psychopharmacology. Rev. Rockville, MD: U.S. National Institute of Health, Psychopharmacology Research Branch, 1976.

Hamilton MA. Rating scale for depression. J Neurol Neurosurg Psychiatry 1960; 23: 56-62. Kawarabayashi T, Younkin LH, Saido TC et al. Age-dependent changes in brain, CSF, and plasma amyloid (beta) protein in the Tg2576 transgenic mouse model of Alzheimer's disease. J Neurosci 2001;15;21(2): 372-381.

Kessing LV, Andersen PK. Does the risk of developing dementia increase with the number of episodes in patients with depressive disorder and in patients with bipolar disorder? J Neurol Neurosurg Psychiatry 2004; 75: 1662-1666.

(12)

Kho KH, Zwinderman AH, Blansjaar BA. Predictors for the efficacy of electroconvulsive therapy: chart review of a naturalistic study. J Clin Psychiatry 2005; 66: 894-899.

Kita Y, Baba H, Maeshima H et al. Serum amyloid beta protein in young and elderly depression: a pilot study. Psychogeriatrics 2009; 9(4): 180-185.

Le Bastard N, Aerts L, Leurs J et al. No correlation between time-linked plasma and CSF Abeta levels. Neurochem Int 2009;55(8): 820-825.

Lecrubier Y. How do you define remission? Acta Psychiatr Scand 2002; 415: 7-11.

Lindefors E, Brodin E, Metsis M. Spatiotemporal selective effects on brain-derived neurotrophic factor and trk B messenger RNA in rat hippocampus by electroconvulsive shock. Neuroscience 1995; 65: 661-670.

Longstreth WT Jr, Manolio TA, Arnold A et al. Clinical correlates of white matter findings on cranial magnetic resonance imaging of 3301 elderly people: the Cardiovascular Health Study. Stroke 1996; 27: 1274-1282.

Marano CM, Phatak P, Vemulapalli UR et al. Increased plasma concentration of brain-derived neurotrophic factor with electroconvulsive therapy: A pilot study in patients with major depression. J Clin Psychiatry 2007; 68: 512-517.

Mehta PD, Pirttila T, Patrick BA et al. Amyloid beta protein 1-40 and 1-42 levels in matched cerebrospinal fluid and plasma from patients with Alzheimer disease. Neurosci Lett 2001;304(1-2): 102-106.

Nibuya M, Morinobu S, Duman RS. Regulation of BDNF and TrkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci 1995; 15: 7539-7547.

Okamoto T, Yoshimura R, Ikenouchi-Sugita A et al. Efficacy of electroconvulsive therapy is associated with changing blood levels of homovanillic acid and brain derived neurotrophic factor (BDNF) in refractory depressed patients: A pilot study. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32: 1185-1190.

Pagnin D, de Queiroz V, Pini S, Cassano GB. Efficacy of ECT in depression: a meta-analytic review. J ECT 2004; 20: 13-20.

Piccinni A, Del Debbio A, Medda P et al. Plasma Brain-Derived Neurotrophic Factor in treatment-resistant depressed patients receiving electroconvulsive therapy. Eur Neuropsychopharmacol 2009; 19(5): 349-355.

Piccinni A, Marazziti D, Catena M et al. Plasma and serum brain-derived neurotrophic factor (BDNF) in depressed patients during 1 year of antidepressant treatments. J Affect Disord 2008; 105: 279-283.

Piccinni A, Origlia N, Veltri A et al. Plasma β–amyloid peptides levels: a pilot study in bipolar depressed patients. J Affect Disord 2012; 138: 160-164.

(13)

Pluijms EM, Birkenhäger TK, Mulder PG, van den Broek WW. Influence of episode duration of major depressive disorder on response to electroconvulsive therapy. J Affect Disord 2006; 90: 233-237.

Pomara N, Murali Doraiswamy P. Does increased platelet release of Abeta peptide contribute to brain abnormalities in individuals with depression? Med Hypotheses 2003;60(5): 640-643. Pomara N, Doraiswamy PM, Willoughby LM et al. Elevation in plasma Abeta42 in geriatric depression: a pilot study. Neurochem Res 2006; 31(3): 341-349.

Pomara N, Sidtis JJ. Brain neurotoxic amyloid-beta peptides: their potential role in the pathophysiology of depression and as molecular therapeutic targets. Br J Pharmacol 2010;161(4): 768-770.

Qiu WQ, Sun X, Selkoe DJ et al. Depression is associated with low plasma Abeta42 independently of cardiovascular disease in the homebound elderly. Int J Geriatr Psychiatry 2007; 22: 536-542.

Robinson LJ, Thompson JM, Gallagher P et al. A meta-analysis of cognitive deficits in euthymic patients with bipolar disorder. J Affect Disord 2006; 93: 105-115.

Selkoe DJ. The ups and downs of Abeta. Nat Med 2006; 12: 758-759.

Sen S, Duman R, Sanacora G. Serum brain-derived neurotrophic factor, depression, and antidepressant medications: Meta-analyses and implications. Biol Psychiatry 2008; 64: 527-532.

Sheehan DV, Lecrubier Y, Sheehan KH et al. The Mini- International Neuropsychiatric Interview (M.I.N.I.): the development and validation of a structured diagnostic psychiatric interview for DSM-IV and ICD-10. J Clin Psychiatry 1998; 59 (Suppl. 20): 22–33.

Sun X, Mwamburi DM, Bungay K et al. Depression, antidepressants, and plasma amyloid beta (beta) peptides in those elderly who do not have cardiovascular disease. Biol Psychiatry 2007; 62: 1413-1417.

Sun X, Steffens DC, Au R et al. Amyloid-associated depression: a prodromal depression of Alzheimer disease? Arch Gen Psychiatry 2008; 65: 542-550.

Taylor SM. Electroconvulsive therapy, brain-derived neurotrophic factor, and possible neurorestorative benefit of the clinical application of electroconvulsive therapy. J ECT 2008; 24: 160-165.

Thase ME, Rush AJ. When at first you don't succeed: sequential strategies for antidepressant nonresponders. J Clin Psychiatry 1997; 58 (Suppl. 13): 23-29.

Tong L, Thornton PL, Balazs R, Cotman CW. Beta-amyloid-(1–42) impairs activity-dependent cAMP-response element-binding protein signaling in neurons at concentrations in which cell survival is not compromised. J Biol Chem 2001; 276: 17301-17306.

Torres IJ, Boudreau VG, Yatham LN. Neuropsychological functioning in euthymic bipolar disorder: a meta-analysis. Acta Psychiatr Scand 2007; Suppl. 434: 17–26.

(14)

van Dijk EJ, Prins ND, Vermeer SE et al. Plasma amyloid beta, apolipoprotein E, lacunar infarcts, and white matter lesions. Ann Neurol 2004; 55: 570-75.

Van Oijen M, Hofman A, Soares HD et al. Plasma Abeta (1-40) and Abeta (1-42) and the risk of dementia: a prospective case cohort study. Lancet Neurol 2006; 5: 655-660.

Vermeer SE, Prins ND, den Heijer T et al. Silent brain infarcts and the risk of dementia and cognitive decline. N Engl J Med 2003; 348: 1215-1222.

Xu W, Kawarabayashi T, Matsubara E et al. Plasma antibodies to Abeta40 and Abeta42 in patients with Alzheimer’s disease and normal controls. Brain Res 2008; 1219: 169-179.

Zhang-Nunes SX, Maat-Schieman ML, van Duinen SG et al. The cerebral beta-amyloid angiopathies: hereditary and sporadic. Brain Pathol 2006; 16: 30-39.

Zimmermann R, Schmitt H, Rotter A et al. Transient increase of plasma concentrations of amyloid β peptides after electroconvulsive therapy. Brain Stimul 2012; 5(1): 25-29.

Zomberg GL, Pope HG. Treatment of depression in bipolar disorder: new directions for research. J Clin Psychopharmacol 1993; 13: 397-408.

Riferimenti

Documenti correlati

Dynamic response of biomass concentration calculated with the virtual plant (continuous line), open-loop model (dashed line) and geometric observer (GO) (dotted grey line) for

L’obiettivo della III Settimana della cultura d’impresa, che si sta tenendo in questi giorni (dal 22 al 28 novembre), organizzata da Confindustria sull’intero territorio nazionale,

Definition of the compressive inelastic strain functional to the definition of the compression hardening data (Abaqus Documentation 2012) ... Key cantilever of the Ridracoli dam

Anaplasmosi e della Febbre Q in Toscana, su campioni di DNA estratti dalle milze prelevate da 100 cinghiali abbattuti nella provincia di Pisa ,durante le stagioni

con la negazione della corporeità del sepolcro stesso (nec molem nec pyrami- dem nec sepulchrum) o più semplicemente con il fatto che il testo dell' enig- ma sia stato

diritti di utilizzazione economica. I Creative Commons condividono con la legge relativa al diritto d’autore e il Codice dei beni culturali e del paesaggio l’obiettivo di

Using the longitudinal and cross-sectional register-trace approaches, we observe less dramatic but still substantive rate ratios for many ages with high fertility intensity, Figure

Two months of verbal opposition by the districts of Berlin, welfare organisations, unions, society for homeless people; foundation of action alliance by welfare organisation