SciELO - Scientific Electronic Library Online

 
vol.31 número6¿Está justificado el tratamiento prolongado con benzodiacepinas?Neuropsychological evaluation in patients with eating disorders índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay artículos similaresSimilares en SciELO

Compartir


Salud mental

versión impresa ISSN 0185-3325

Salud Ment vol.31 no.6 México nov./dic. 2008

 

Conferencia magistral

 

Genetic studies of bipolar disorder in patients selected by their treatment response*

 

Abigail Ortiz–Domínguez,1 Martin Alda*1

 

1 Department of Psychiatry, Dalhousie University.

 

*Correspondence:
Dr. Martin Alda.
Department of Psychiatry,Dalhousie University.
5909 Veterans' Memorial Lane,
Halifax, Nova Scotia,Canada,B3H 3E2.
Fax number: + (902) 4734877
Email:
malda@dal.ca

 

Abstract

Bipolar disorder (BD) is a major mood disorder with several genes of moderate or small effect contributing to the genetic susceptibility. It is also likely heterogeneous, which stimulated efforts to refine its clinical phenotype, studies investigating the link between BD susceptibility and response to a specific mood stabilizer appear to be one of the promising directions.

In particular, excellent response to lithium prophylaxis has been described as a clinical marker of a more homogeneous subgroup of BD, characterized by an episodic course, low rates of co–morbid conditions, absence of rapid cycling, and a strong genetic loading. These results also suggest that lithium response clusters in families (independent of the increased familial loading for affective disorders), likely on a genetic basis.

For almost 40 years, clinical studies have pointed to differences between lithium responders (LR) and non–responders (LNR). For instance, there is a higher frequency of BD in LR families. As well, investigations in offspring of LR and LNR probands show that the offspring of LR tend to manifest a higher frequency of affective disorders, less co–morbidity and an episodic course of the disorder, compared with the offspring of LNR, who had a broad range of psychopathology, a higher rate of co–morbidity and a chronic course of the disorder.

A number of candidate genes have been studied in patients treated with lithium; of these, several showed an association in at least one study: cAMP responsive element binding protein (CREB), X–box binding protein 1 (XBP–1), inositol polyphosphate–1–phosphatase (INNP1), serotonin transporter gene (5–HTT), brain–derived neurotrophic factor (BDNF), phospholipase γ–1 (PLCγ–1), dopamine receptors (D2 and D4), polyglutamine tracts, tyrosine hydroxylase, inositol monophosphatase (IMPA), mitochondrial DNA, and breakpoint cluster region (BCR) gene.

Clinical studies have shown as well that the treatment response and outcome appear to be specific for the different types of mood stabilizers. Patients who respond to lithium exhibit qualitative differences from patients responding to other medications, such as valproate, carbamazepine or lamotrigine. Responders to carbamazepine had atypical clinical features, such as mood–incongruent psychosis, an age at onset of illness below 30 years old, and a negative family history of mood disorders. Similarly, in a study comparing the phenotypic spectra in responders to lithium versus lamotrigine the probands differed with respect to clinical course (with rapid cycling and non–episodic course in the lamotrigine group) and co–morbidity, with the lamotrigine–responder group showing a higher frequency of panic attacks and substance abuse.

In conclusion, pharmacogenetic studies may provide important clues to the nature of bipolar disorder and the response to long term treatment.

Key words: Lithium response, pharmacogenetic, probands.

 

Resumen

El trastorno bipolar (TB) es un trastorno afectivo con varios genes, de efecto leve o moderado, que contribuyen a su susceptibilidad. Es asimismo un trastorno heterogéneo, lo que ha estimulado diversas iniciativas para refinar el fenotipo de los pacientes con este trastorno. Particularmente en el TB, una respuesta excelente a la profilaxis con litio ha sido descrita como un marcador clínico en un subgrupo más homogéneo en TB, caracterizado por un curso episódico, baja prevalencia respecto a comorbilidad, ausencia de ciclado rápido y una carga genética importante. En relación con ello, y a pesar de que la totalidad de los estudios no coinciden, la mayor parte sugiere que seleccionar <<probandos>> de acuerdo con su respuesta al tratamiento incrementa la homogeneidad fenotípica. Estos resultados sugieren asimismo que la respuesta al litio <<se agrupa>> en familias (independientemente de la tasa familiar incrementada para trastornos afectivos), muy probablemente con bases genéticas.

Por casi 40 años, los estudios clínicos han dilucidado las diferencias entre los respondedores a litio (LR) y los no respondedores (LNR). A este respecto, existe una frecuencia más alta de TB en familias LR; asimismo, las investigaciones en los descendientes de los probandos LR y LNR han demostrado que los descendientes de LR tienden a manifestar una mayor frecuencia de trastornos afectivos, menor comorbilidad, y un curso episódico del trastorno comparados con los descendientes de LNR, quienes muestran un amplio espectro de psicopatología, una alta tasa de comorbilidad, y un curso crónico del trastorno.

Diversos genes candidatos han sido estudiados en pacientes tratados con litio, y varios de ellos han mostrado una asociación en al menos un estudio: proteína de unión al elemento de respuesta (cAMP responsive element binding protein, CREB), proteína de unión a X–box 1 (X–box binding protein 1, XBP–1), inositol polifosfato–1–fosfatasa (INNP1), transportador de serotonina (5–HTT), factor de crecimiento derivado del cerebro (brain–derived neurotrophic factor, BDNF), fosfolipasa γ–1(PLCγ–1), receptores dopaminérgicos (D2 y D4), poliglutamina, tirosina hidroxilasa, inositol monofosfatasa (IMPA), DNA mitocondrial y el gen BCR.

Los estudios clínicos han demostrado que la respuesta al tratamiento y el pronóstico parecen ser específicos para los diferentes tipos de estabilizadores del ánimo. Los pacientes que responden al litio exhiben diferencias cualitativas respecto a los pacientes que responden a otros estabilizadores del ánimo, como valproato, carbamazepina o lamotrigina. Los respondedores a carbamazepina presentan características clínicas atípicas, como psicosis incongruente con el afecto, una edad de inicio del trastorno menor a 30 años de edad y no cuentan con historia familiar de trastornos afectivos. Finalmente, un estudio entre los respondedores al litio y los respondedores a lamotrigina demostró que los probandos difieren con respecto al curso clínico del trastorno (con ciclado rápido y un curso no episódico en los pacientes que responden a lamotrigina), así como en la comorbilidad, teniendo los pacientes que responden a lamotrigina una mayor frecuencia de crisis de angustia y abuso de sustancias.

En conclusión, los estudios farmacogenéticos podrían proveer de hallazgos importantes respecto a la naturaleza del trastorno bipolar y la respuesta al tratamiento a largo plazo.

Palabras clave: Respuesta al litio, farmacogenética, probandos.

 

GENETIC STUDIES OF BIPOLAR DISORDER IN PATIENTS SELECTED BY THEIR TREATMENT RESPONSE

Bipolar disorder (BD) is a major mood disorder with an important genetic contribution to its etiology.1,2 The currently accepted genetic model of BD is that of oligogenic inheritance with several genes of moderate or small effects contributing to the genetic susceptibility. Some of the chromosomal regions supported by more than one study are those in 4p, 12q24, 13q, 18p11, 18q and 22q.3,4 However, many of these findings are not consistent for a variety of reasons, including methodological differences, ascertainment schemes, and markers genotyped.

Bipolar disorder is likely heterogeneous, which stimulated efforts to refine its clinical phenotype. These include classification of affected subjects based on their clinical characteristics, such as the presence of co–morbid conditions, specific symptoms, especially psychosis, and response to treatment. In particular, studies investigating the link between BD susceptibility and response to a specific mood stabilizer appear to be a promising direction; hence, the focus of this review will be to describe the pharmacogenetic characteristics associated with a specific response to a mood stabilizer.

 

LITHIUM

Phenotypic characterization of responders to lithium

Excellent response to lithium prophylaxis has been described as a clinical marker of a more homogeneous subgroup of BD, characterized by an episodic course,5 low rates of co–morbid conditions,3 absence of rapid cycling,6 and a strong genetic loading.7–9 Not all, but most studies of lithium response suggest that selecting probands according to treatment outcome increases phenotypic homogeneity. These results also suggest that lithium response clusters in families (independent of the increased familial loading for affective disorders), likely on a genetic basis.10 Likewise, several studies elucidated the increased rates of affective disorders among relatives of primary affective disorder probands using the outcome on lithium treatment as a discriminatory criterion.11,12 Several reports indicated also a relatively simple model of inheritance, in some cases compatible with a major–gene effect in lithium responders (LR).8,9

For almost 40 years, clinical studies have pointed to differences between LR and non–responders (LNR), with reproducible findings in LR probands and their relatives, as described in table 1. Although clinical methods to evaluate lithium response differ between research groups, findings paint a consistent picture. Specifically, there is a higher frequency of BD in LR families,11,13–15 as well as a higher prevalence of unequivocal response to lithium in relatives of LR probands;16–18 conversely, a higher frequency of schizophrenic spectrum disorders has been reported in the LNR families.19,20 Other studies have as well reported the association between the episode sequence and lithium response. Specifically, a predominant pattern of mania–depression–free interval (MDI) has been positively associated with lithium response.6,21

However, family history studies are not unequivocal; some could not find any difference between lithium responders and non–responders22,23 found an inverse association between family history and lithium response.24,25 The smaller number of discrepant findings, as pointed by Alda,26 could be related to different methodologies, particularly in the definition of lithium response and non–response, and the changing trends in the diagnosis of mood disorders.27

Studies in offspring of bipolar parents and lithium response

Investigations in offspring of LR and LNR probands are consistent with family and family history studies. More than 20 years ago, McKnew's28 findings on the offspring of BD patients supported the association between lithium response in the parents and in the offspring. More recent studies have confirmed these findings; for instance, Duffy and colleagues29 studied the offspring of 13 LR probands (n=21), and the offspring of 10 LNR probands (n=15). Her findings show that the offspring of LR tend to manifest a higher frequency of affective disorders, less co–morbidity, and an episodic course of the disorder, compared with the offspring of LNR, who had a broad range of psychopathology, a higher rate of co–morbidity, and a chronic course of the disorder. A prospective study in this high–risk population showed as well that among the LR offspring there was no occurrence of schizoaffective disorders, and confirmed the association between parental course of the disorder and lithium response in the offspring.30 In addition, another study from the same group suggested that treatment response in the affected offspring is associated with treatment response in the affected parent.31 In summary, family studies suggest that LR are genetically distinct from LNR, and that their disorder appears to have a stronger genetic basis,32 findings that have also been supported by studies of age at onset and LR24,33 (Ortiz et al., in preparation). Nevertheless, the response to treatment could be influenced by a separate genetic factor, independent of the predisposition to the illness itself, as suggested from the findings of Turecki, hypothesizing that the locus on chromosome 15 may be involved in the etiology of BD, whereas the locus on chromosome 7 may be related to the response to lithium treatment.34

Molecular genetic studies in patients treated with lithium

A number of studies examined specific candidate genes and response to lithium; typically, these genes have been selected based on possible mechanism of action. However, most of the pharmacogenetic studies have yielded conflicting results (table 2).

a) cAMP responsive element binding protein (CREB): A transcription factor that increases the expression of key growth factors involved in synaptogenesis and neurogenesis, has been proposed as a target in the study of BD, due to its role in gene expression. An association study by Mamdani et al35 compared 180 LR, 69 LNR, and 127 controls. The researchers reported an association between lithium response and CREB1–1H SNP (G/A change) and the CREB1–7H SNP (T/C change). The CREB1 gene has as well been associated to recurrent depression in women suggesting the existence of a sex–specific susceptibility gene that could interact with nuclear estrogen receptors.36,37

b) X–box binding protein 1 (XBP–1): The transcription factor X–box binding protein (XBP–1) was first identified by its ability to bind to the x–box, a conserved transcriptional element in the human leukocyte antigen (HLA) DR alpha promoter. XBP–1 is upregulated as part of the endoplasmic reticulum (ER) stress response. It has been found recently that the 116CG polymorphism causes an impairment of the ER stress response and increases the risk of BD,38 while there have been reports regarding the association between the –116C/G SNP pf the XBP1 gene and lithium prophylaxis in BD.39 Taken together, these studies suggest the association of a lower XBP1 expression and lack of response to lithium.26

c) Inositol polyphosphate–1–phosphatase (INNP1): INPP1 encodes the enzyme inositol polyphosphate–1–phosphatase, one of the enzymes involved in phosphatidylinositol signaling pathways, which has been studied with respect to the therapeutic action of lithium. In fact, the study from Steen40 in a Norwegian sample showed that 67% of lithium responders showed the C937A polymorphism, compared with 11% of non–responders, although some other studies have not replicated these findings.41

d) Serotonin transporter gene (5–HTT): One of the most studied genes of BD,42,43 being the most commonly investigated polymorphism the insertion/deletion polymorphism in the promoter region. With regard to lithium response, conflicting results have been obtained, in the sense that previous studies have reported that the short allele variant is associated with a poor response to lithium,44–46 although other studies have indicated the opposite association.47

e) Brain–derived neurotrophic factor (BDNF): BDNF is a neurotrophic factor implicated in neuronal proliferation and synaptic plasticity. Conflicting results regarding an association between BD and BDNF have been reported,48,49 although in other studies rapid cycling has been associated with Val66met polymorphism of BDNF.50 In lithium responders, Rybakowski et al.51 studied 88 BD patients, stratified by lithium response as excellent, partial or non–responders. His findings support the association of Val/Met genotype of Val66Met polymorphism with excellent responders, as well as a possible interaction between this genotype and the serotonin transporter in lithium responders,52 although studies with larger samples have contradicted these initial findings.53 This could probably be related to differences in the ascertainment of lithium response.

f) Phospholipase γ–1 (PLCγ–1): Lithium is thought to stabilize mood by acting at the phosphoinositide cycle, and the γ–1 isozyme of phospholipase C plays an important role in the phosphoinositide second messenger system. Therefore, polymorphisms in the PLCγ–1 gene have been investigated in lithium responders. The study by Turecki et al.54 involved over 130 patients, and found an association between lithium response and the PLC γ–1/ 5 polymorphism. Although his findings have been replicated by other groups,55 other studies have failed to replicate the initial findings.56

g) Dopamine receptors (DR) D2 and D4: The DRD2 gene, localized at 11q22.3–q23,57 and the DRD4 gene,58 located near the telomere of chromosome 11p,59 are candidates for prediction of lithium response in mood disorders. However, most studies in lithium–responsive patients have not confirmed the initial positive findings.60,61

h) Polyglutamine tracts: Family studies showing anticipation and RED studies indicating that bipolar patients have longer CAG repeats have suggested that trinucleotide repeats may play a role in the etiology of gp 62–64 However, a recent replication study from previous findings did not support the association with triple repeat expansion in ERDA1 and CTG18.1.65 Two studies involving lithium–responsive patients did not confirmed either the association.66,67

i) Tyrosine hydroxylase (TH): Tyrosine hydroxylase is an oxidase that converts tyrosine to L–hydroxyphenyl–alanine (L–DOPA). This enzyme is rate–limiting for the synthesis of both dopamine and norepinephrine; and the TH gene has been localized to the short arm of chromosome 11 (11p15). The cumulative evidence available to date regarding linkage studies of chromosome 11 markers and bipolar disorder report no association between the TH gene and BD.6871

j) Inositol monophosphatase (IMPA): The activity of IMPA, the target enzyme of lithium in the phosphatidilinositol (PI) signal transduction system, has been another candidate for the study of lithium response, and studies have shown a lower activity of IMPA in cells lines from lithium–responsive patients.72 Two genes coding for IMPA, IMPA1 and IMPA2 have been identified; the first is localized to chromosome 8q21.13–21.373 and the second in the chromosomal region 18p11.2. Whereas no linkage or association studies have provided so far evidence for a role of IMPA1 in BD, there have been many reports suggesting the role of 18p11.2 as a susceptibility locus for the disorder;74–77 none of the studies investigated patients selected by their lithium response, though. There are two association studies that did included this criteria: the one from Sjoholt,78 which reports an apparent significant association between IMPA2 and BD; however, the one from Dimitrova79 did not find any significant association between LR and IMPA2.

k) Mitochondrial DNA: Mitochondrial dysfunction has been implicated in the physiopathology of BD, based in findings related to abnormal brain energy metabolism, increased levels of 4977–bp deletion in mitochondrial DNA, and co–morbidity of affective disorders in certain types of mitochondrial disorders;80 the association of mitochondrial DNA (mtDNA) 5178 and 10398 polymorphisms with BD has been reported as well.81 Recently, in a study involving 34 lithium responders and 20 non–responders, Washizuka reported the association between mtDNA 10389A polymorphism and lithium response;82 however, more studies are needed to confirm this association.

l) Other candidate genes: Having conducted a complete genome scan in 247 individuals, the study from Turecki et al.34 suggested that a locus on chromosome 15 (15q 14) may be involved in the etiology of the disorder, whereas the locus on chromosome 7 (7q1 1.2) may be involved in lithium response.

A recent study from Masui83 has as well implicated the breakpoint cluster region gene (BCR), which is located on chromosome 22q11, and lithium response. The study, that included 43 LR and 118 LNR, showed that lithium treatment might be less effective in patients homozygous for the Ser796 allele of the BCR gene than in patients with the Asn796 allele; in addition, it showed that the same variant associated with the illness was associated with a poorer outcome, which raises the possibility of an association between BCR Ser796 and a more severe illness presentation.

Although most of the following studies have not been replicated, the following gene variants did not appear to be associated with lithium outcome in BD: MAO A,84,85 TPH,86 Gamma–Aminobutyric Acid (GABA) receptors (A3, A5, B3),87 serotonin receptors (5HT2A, C, and 1A),88 CRH and PENK genes,89 Catechol–O–methyl transferase (COMT),84 Glycogen synthase kinase 3 (GSK–3β),90 and PREP.91

 

OTHER MOOD STABILIZERS

Clinical studies have shown that the treatment response and outcome appear to be specific for the different types of mood stabilizers. Specifically, patients who respond to lithium exhibit qualitative differences from patients responding to other medications, such as valproate, carbamazepine or lamotrigine.3 In addition, treatment response could be a specific characteristic of individual patients, i.e., patients responding to carbamazepine or valproate had evidence of prior nonresponse to lithium.92,93

Using positron emission tomography, Kruger and colleagues94 analyzed the changes in regional cerebral blood flow (rCBF) after induction of transient sadness in euthymic LR (n=9) and nine healthy siblings, and compared them to the results from nine valproate–responsive bipolar patients. Among their findings, the group reports changes specific to the bipolar subgroup in the dorsolateral prefrontal cortex and rostral anterior cingulated that distinguished valproate–from lithium–responsive patients. Also, the results yielded an insight on the changes in the medial frontal cortex that distinguished both lithium– and valproate–responsive patients from healthy siblings.

 

VALPROATE

Several studies suggest common biochemical pathways for lithium and valproate, including the extracelullar signal–regulated kinase systems;95 particularly for valproate, alterations in the Na+ channel subunit mRNA levels and MARCKS expression have been reported.96

Valproate has been effective in the prophylaxis of rapid cycling and ultra–rapid cycling bipolar disorders; as well as in the acute treatment of mania and mixed states.93,97,98 Other studies have reported a good antimanic response to valproate associated with decreasing or stable episode frequencies, and nonpsychotic mania.99 However, in a naturalistic observation study that included 120 BD patients, responders to valproate had higher rates of psychosis.100 Among other variables studied, a recent study from Reeves101 including 20 participants (6 lithium–responsive and 14 valproate–responsive) showed that specific EEG abnormalities, such as sharp activity, could predict response to valproate.

Molecular studies with valproate, although scarce in comparison with those including lithium, have started to show the implications of genes essential in the endoplasmic reticulum (ER) stress response signalling. For instance, the study from Kakiuchi et al.38 used DNA microarray analysis of lymphoblastoid cells derived from two pairs of twins discordant with respect to BD, and they found downregulated expression of genes related to the ER response in both affected twins. The study shows thus that the 116CG SNP of X–box binding protein 1 (XPBP–1) causes an impairment of the ER stress response, and was significantly associated with BD. Their results illustrate as well the differential effect of the mood stabilizers on this cascade, specifically, that valproate administration significantly ameliorates the ER stress response compromised by the risk allele –116G by reinforcing ATF6 upstream of the XBP1 loop, while lithium and carbamazepine did not.

 

CARBAMAZEPINE

Clinical studies have reported that, unlike lithium responders, patients who respond to carbamazepine had atypical clinical features, such as mood–incongruent psychosis;102 as well as an age at onset of illness below 30 years old.33 Furthermore, rapid cycling and a negative family history of mood disorders may be associated with a good response to carbamazepine.92

The multicenter Study of Long Term Treatment of Affective and Schizoaffective Psychosis (MAP study) was a prospective study with an observation period of more than two years, trying to compare the differential efficacy of lithium and carbamazepine.103 The study included 171 BD patients: 86 were randomized to lithium and 85 to carbamazepine. All data consistently suggested that lithium was more efficacious than carbamazepine in the maintenance treatment of BD 1 patients, and in those patients with <<classical features>>, i.e., bipolar I patients without mood–incongruent delusions and without psychiatric co–morbidity.

On the contrary, patients with non–classical features responded more favourably to carbamazepine, as there was an inverse association between the number of non–classical features and hospitalization rate for patients under treatment with carbamazepine.

 

LAMOTRIGINE

A study comparing the phenotypic spectra in responders to lithium versus lamotrigine showed that the probands differed with respect to clinical course (with rapid cycling and non–episodic course in the lamotrigine group) and co–morbidity, having the lamotrigine–responder group a higher frequency of panic attacks and substance abuse. Family history was also an important predictor, that is, relatives of lamotrigine responders had a higher prevalence of schizoaffective disorder, major depression, and panic attacks.104 Other studies comparing lamotrigine and gabapentin have found that lamotrigine appeared more effective in patients with fewer medication trials, whereas gabapentin appeared most effective in those with younger age and lower baseline weight.105

 

CONCLUSIONS

Recent progress in pharmacogenetic studies has revealed the promissory future of genetic studies according to treatment response, providing a better framework for the understanding of bipolar disorder. However, the main challenge in this realm comprises the integration of genetic, molecular, cognitive and clinical domains, in order to refine the study of the broad spectrum of clinical presentations that can be found in bipolar disorder. Treatment response appears as an invaluable tool for a better understanding of such a complex disorder.

 

REFERENCES

1. Baron M. Manic–depression genes and the new millenium: poised for discovery. Molecular Psychiatry 2002;7(4):342–358.        [ Links ]

2. Segurado R, Tera–Wadleigh S, Levinson D. Genome scan meta–analysis of schizophrenia and bipolar disorder, part III: Bipolar disorder. Am J Med Genet 2003;73(1):49–62.        [ Links ]

3. Alda M, Grof P, Rouleau G, Turecki G, Trevor Young L. Investigating responders to lithium prophylaxis as a strategey for mapping susceptibility genes for Bipolar Disorder. Prog Neuropsychopharmacol Biol Psychiatry 2005;29(6):1038–1045.        [ Links ]

4. Badner J, Gershon E. Meta–analysis of whole–genome linkage scans of bipolar disorder and schizophrenia. Molecular Psychiatry 2002;7(4):405–11.        [ Links ]

5. Alda M. Genetic factors and treatment of mood disorders. Bipolar Disorders 2001;3:318–324.        [ Links ]

6. Kleindienst N, Engel R, Greil W. Which clinical factors predict response to prophylactic lithium? A systematic review for bipolar disorders. Bipolar Disorders 2005;7:404–417.        [ Links ]

7. MacQueen G, Hajek T, Alda M. The phenotypes of bipolar disorder: relevance for genetic investigations. Molecular Psychiatry 2005;10:811–826.        [ Links ]

8. Alda M, Grof P, Grof E, Zvolsky P, Walsh M. Mode of inheritance in families of patients with lithium–responsive affective disorders. Acta Psychiatr Scand 1994;90:304–310.        [ Links ]

9. Alda M, Grof E, Cavazzoni P, Duffy A, Martin R et al. Autosomal recessive inheritance of affective disorders in families of responders to lithium prophylaxis? J Affective Disorders 1997;44:153–157.        [ Links ]

10. Mamdani F, Jaitovich Groisman I, Alda M, Turecki G. Pharmacogenetics and bipolar disorder. Pharmacogenom J 2004;4:161–170.        [ Links ]

11. Smeraldi E, Petroccione A, Gasperini M, Macciardi F, Orsini A et al. Outcomes on lithium treatment as a tool for genetic studies in affective disorders. J Affective Disorders 1984;6:139–151.        [ Links ]

12. Smeraldi E, Petroccione A, Gasperini M, Macciardi F, Orsini A. The search for Genetic Homogeneity in Affective Disorders. J Affective Disorders 1984;7:99–107.        [ Links ]

13. Abou–Saleh MT, Coppen A. Who responds to prophylactic lithium? J Affect Disord 1986; 10(2): 115–125.        [ Links ]

14. Grof P, Alda M, Grof E, Zvolsky P, Walsh M. Lithium response and genetics of affective disorders. J Affective Disorders 1994;32:85–95.        [ Links ]

15. Mendlewicz J, Fieve R, Stallone F. Relationship between the efectiveness of lithium therapy and family history. American J Psychiatry 1973;130(9):1011–1013.        [ Links ]

16. Grof P, Duffy A, Cavazzoni P et al. Is response to prophylactic lithium a familial trait? J Clinical Psychiatry 2002;63(10) :942–947.        [ Links ]

17. Maj M, Del VM, Starace F, Pirozzi R, Kemali D. Prediction of affective psychoses response to lithium prophylaxis. The role of socio–demographic, clinical, psychological and biological variables. Acta Psychiatr Scand 1984; 69(1):37–44.        [ Links ]

18. Mendlewicz J, Fieve RR, Stallone F, Fleiss JL. Genetic history as a predictor of lithium response in manic–depressive illness. Lancet 1972; 11;1(7750):599–600.        [ Links ]

19. Aronoff MS, Epstein RS. Factors associated with poor response to lithium carbonate: a clinical study. Am J Psychiatry 1970;127(4) :472–480.        [ Links ]

20. Sautter F, Garver D. Familial differences in lithium response versus lithium nonresponse psychoses. J Psychiatric Research 1985;19(1):1–8.        [ Links ]

21. Maj M, Pirozzi R, Starace F. Previous pattern of course of the illness as a predictor of response to lithium prophylaxis in bipolar patients. J Affect Disord 1989; 17(3) :237–241.        [ Links ]

22. Coryell W, Akiskal H, Leon A, Solomon D, Endicott J. Family history and symptom levels during treatment for bipolar I affective disorder. Biological Psychiatry 2000;47:1034–1042.        [ Links ]

23. Dunner DL, Fleiss JL, Fieve RR. Lithium carbonate prophylaxis failure. Br J Psychiatry 1976y;129:40–44.        [ Links ]

24. Engstrom C, Astrom M, Nordqvist K, Adolfsson R, Nylander P. Relationship between prophylactic effect of lithium therapy and family history of affective disorders. Biological Psychiatry 1997;42:425–433.        [ Links ]

25. Misra PC, Burns BH. <<Lithium non–responders>> in a lithium clinic. Acta Psychiatr Scand 1977;55(1):32–40.        [ Links ]

26. Alda M, Grof P, Zvolsky P. Genetic factors and response to lithium treatment. Ref Type: Serial (Book,Monograph) 2008.        [ Links ]

27. Grof P, Alda M. Discrepancies in the efficacy of lithium. Archives General Psychiatry 2000;57:191.        [ Links ]

28. McKnew DH, Cytryn L, Buchsbaum MS et al. Lithium in children of lithium–responding parents. Psychiatry Res 1981 ;4(2): 171–180.        [ Links ]

29. Duffy A, Alda M, Kutcher S, Fusee C, Grof P. Psychiatric symptoms and syndromes among adolescent children of parents with lithium–responsive or lithium–nonresponsive bipolar disorder. American J Psychiatry 1998;155(3):431–433.        [ Links ]

30. Duffy A, Alda M, Kutcher S et al. A prospective study of the offspring of bipolar parents responsive and nonresponsive to lithium treatment. J Clinical Psychiatry 2002;63(12):1 171–1178.        [ Links ]

31. Duffy A, Alda M, Milin R, Grof P. A consecutive series of treated affected offspring of parents with bipolar disorder: is response associated with the clinical profile? Canadian J Psychiatry 2007;52(6) :369–376.        [ Links ]

32. Alda M. Pharmacogenetics of lithium response in bipolar disorder. J Psychiatry Neuroscience 1999;24(2):154–158.        [ Links ]

33. Okuma T. Effects of carbamazepine and lithium on affective disorders. Neuropsychobiology 1993;27:138–145.        [ Links ]

34. Turecki G, Grof P, Grof E et al. Mapping susceptibility genes for bipolar disorder: a pharmacogenetic approach based on excellent response to lithium. Molecular Psychiatry 2001;6:570–578.        [ Links ]

35. Mamdani F, Alda M, Grof P, Trevor Young L, Rouleau G et al. Lithium response and genetic variation in the CREB family of genes. Am J Med Genet Part B 2008.        [ Links ]

36. Zubenko GS, Hughes HB, III, Maher BS, Stiffler JS, Zubenko WN et al. Genetic linkage of region containing the CREB1 gene to depressive disorders in women from families with recurrent, early–onset, major depression. Am J Med Genet 2002; 114(8) :980–987.        [ Links ]

37. Zubenko GS, Hughes HB, III, Stiffler JS et al. Sequence variations in CREB1 cosegregate with depressive disorders in women. Mol Psychiatry 2003;8(6):611–618.        [ Links ]

38. Kakiuchi C, Iwamoto K, Ishiwata M et al. Impaired feedback regulation of XBP1 as a genetic risk factor for bipolar disorder. Nature Genetics 2003;35(2):171–175.        [ Links ]

39. Masui T, Hashimoto R, Kusumi I et al. A possible association between the –116 C/G single nucleotide polymorphism of the XBP1 gene and lithium prophylaxis in bipolar disorder. International J Neuropsycho–pharmacology 2006;9:83–88.        [ Links ]

40. Steen V, Lovlie R, Osher Y, Belmaker R, Berle J et al. The polymorphic inositol polyphosphate 1–phosphatase gene as a candidate for pharmacogenetic prediction of lithium–responsive manic–depressive illness. Pharmacogenetics 1998;8:259–268.        [ Links ]

41. Michelon L, Meria–Lima I, Cordeiro Q et al. Association study of the INPP–1, 5HTT, BDNF, AP–2â, and GSK–3â GENE variants and retrospectively scored response to lithium prophylaxis in bipolar disorder. Neuroscience Letters 2006;403:288–293.        [ Links ]

42. Kato T. Molecular genetics of bipolar disorder. Neurosci Res 2001;40(2): 105–113.        [ Links ]

43. Kato T. Molecular genetics of bipolar disorder and depression. Psychiatry Clin Neurosci 2007;61(1):3–19.        [ Links ]

44. Rybakowski J, Suwalska A, Czerski P, Dmitrzak–Weglarz M, Leszcynska–Rodziewicz A et al. Prophylactic effect of lithium in bipolar affective illness may be related to serotonin transporter genotype. Pharmacological Reports 2005;57:124–127.        [ Links ]

45. Serretti A, Lilli R, Mandelli L, Lorenzi C, Smeraldi E. Serotonin transporter gene associated with lithium prophylaxis in mood disorders. Pharmacogenom J 2001;1:71–77.        [ Links ]

46. Ikeda A, Kato T. Biological predictors of lithium response in bipolar disorder. Psychiatry Clinical Neurosciences 2003;57:243–250.        [ Links ]

47. Del Zompo M, Ardau R, Palmas M, Bocchetta A, Reina A et al. Lithium response: association study with two candidate genes. Molecular Psychiatry 1999;4:S66.        [ Links ]

48. Kunugi H, Iijima Y, Tatsumi M et al. No association between the Val66Met polymorphism of the brain–derived neurotrophic factor gene and bipolar disorder in a Japanese population: a multicenter study. Biol Psychiatry 2004; 1;56(5):376–378.        [ Links ]

49. Lohoff FW, Sander T, Ferraro TN, Dahl JP, Gallinat J et al. Confirmation of association between the Val66Met polymorphism in the brain–derived neurotrophic factor (BDNF) gene and bipolar I disorder. Am J Med Genet B Neuropsychiatr Genet 2005;139(1):51–53.        [ Links ]

50. Green EK, Raybould R, Macgregor S et al. Genetic variation of brain–derived neurotrophic factor (BDNF) in bipolar disorder: case–control study of over 3000 individuals from the UK. Br J Psychiatry 2006;188:21–25.        [ Links ]

51. Rybakowski J, Suwalska A, Skibinska M et al. Prophylactic lithium response and polymorphysm of the brain–derived neurotrophic factor gene. Pharmacopsychiatry 2005;38:166–170.        [ Links ]

52. Rybakowski JK, Suwalska A, Skibinska M, Dmitrzak–Weglarz M, Leszczynska–Rodziewicz A et al. Response to lithium prophylaxis: interaction between serotonin transporter and BDNF genes. Am J Med Genet B Neuropsychiatr Genet 2007;144(6):820–823.        [ Links ]

53. Masui T, Hashimoto R, Kusumi I et al. Lithium response and Val66Met polymorphism of the brain–derived neurotrophic factor gene in Japanese patients with bipolar disorder. Psychiatr Genet 2006; 16(2):49–50.        [ Links ]

54. Turecki G, Grof P, Cavazzoni P, Duffy A, Grof E et al. Evidence for a role of phospholypase C – gamma 1 in the pathogenesis of bipolar disorder. Molecular Psychiatry 1998;3:534–538.        [ Links ]

55. Lovlie R, Berle J, Stordal E, Steen V. The phospoliphase C–gamma 1 gene (PLCG1) and lithium–responsive bipolar disorder: re–examination of an intronic dinucleotide repeat polymorphism. Psychiatric Genetics 2001;11:41–43.        [ Links ]

56. Ftouhi–Paquin N, Alda M, Grof P, Chretien N, Rouleau G et al. Identification of three polymorphisms in the translated region of PLC–gamma 1 and their investigation in lithium responsive bipolar disorder. Am J Med Genet 2001;105:301–305.        [ Links ]

57. Grandy DK, Litt M, Allen L et al. The human dopamine D2 receptor gene is located on chromosome 11 at q22–q23 and identifies a TaqI RFLP. Am J Hum Genet 1989;45(5) :778–785.        [ Links ]

58. Van Tol HH, Wu CM, Guan HC et al. Multiple dopamine D4 receptor variants in the human population. Nature 1992;358(6382): 149–152.        [ Links ]

59. Wang E, Ding YC, Flodman P et al. The genetic architecture of selection at the human dopamine receptor D4 (DRD4) gene locus. Am J Hum Genet 2004;74(5):931–944.        [ Links ]

60. Serretti A, Lilli R, Lorenzi C, Franchini L, Smeraldi E. Dopamine receptor D3 gene and response to lithium prophylaxis in mood disorders. International J Neuropsychopharmacology 1998;1:125–129.        [ Links ]

61. Serretti A, Lilli R, Lorenzi C, Gasperini M, Smeraldi E. Dopamine receptor D2 and D4 genes, GABA (A) alpha–1 subunit genes and response to lithium prophylaxis in mood disorders. Psychiatry Research 1999;87:7–19.        [ Links ]

62. McInnis MG, McMahon FJ, Chase GA, Simpson SG, Ross CA et al. Anticipation in bipolar affective disorder. Am J Hum Genet 1993;53(2) :385–390.        [ Links ]

63. O'Donovan MC, Guy C, Craddock N et al. Expanded CAG repeats in schizophrenia and bipolar disorder. Nat Genet 1995;10(4) :380–381.        [ Links ]

64. Mendlewicz J, Lindbald K, Souery D et al. Expanded trinucleotide CAG repeats in families with bipolar affective disorder. Biol Psychiatry 1997;42(12):1115–1122.        [ Links ]

65. Mendlewicz J, Souery D, Del–Favero J et al. Expanded RED products and loci containing CAG/CTG repeats on chromosome 17 (ERDA1) and chromosome 18 (CTG18.1) in trans–generational pairs with bipolar affective disorder. Am J Med Genet B Neuropsychiatr Genet 2004;128(1) :71–75.        [ Links ]

66. Turecki G, Alda M, Grof P et al. Polyglutamine tracts: no evidence of a major role in bipolar disorder. Mol Psychiatry 1999;4(3) :220–221.        [ Links ]

67. Turecki G, Alda M, Grof P et al. Polyglutamine coding genes in bipolar disorder: lack of association with selected candidate loci. J Affect Disord 2000;58(1):63–68.        [ Links ]

68. De BA, Mendelbaum K, Sandkuijl LA et al. Nonlinkage of bipolar illness to tyrosine hydroxylase, tyrosinase, and D2 and D4 dopamine receptor genes on chromosome 11. Am J Psychiatry 1994;151(1) :102–106.        [ Links ]

69. Mendlewicz J, Leboyer M, De BA et al. Absence of linkage between chromosome 11p15 markers and manic–depressive illness in a Belgian pedigree. Am J Psychiatry 1991; 148(12) :1683–1687.        [ Links ]

70. Mitchell P, Waters B, Morrison N, Shine J, Donald J et al. Close linkage of bipolar disorder to chromosome 11 markers is excluded in two large Australian pedigrees. J Affect Disord 1991;21 (1):23–32.        [ Links ]

71. Cavazzoni P, Alda M, Turecki G et al. Lithium–responsive affective disorders: no association with the tyrosine–hydroxilase gene. Psychiatry Research 1996;64:91–96.        [ Links ]

72. Shamir A, Ebstein RP, Nemanov L, Zohar A, Belmaker RH et al. Inositol monophosphatase in immortalized lymphoblastoid cell lines indicates susceptibility to bipolar disorder and response to lithium therapy. Mol Psychiatry 1998;3(6) :481–482.        [ Links ]

73. Sjoholt G, Molven A, Lovlie R, Wilcox A, Sikela JM et al. Genomic structure and chromosomal localization of a human myo–inositol monophosphatase gene (IMPA). Genomics 1997;45(1):113–122.        [ Links ]

74. Berrettini WH, Ferraro TN, Goldin LR et al. Chromosome 18 DNA markers and manic–depressive illness: evidence for a susceptibility gene. Proc Natl Acad Sci USA 1994;91 (13):5918–5921.        [ Links ]

75. Detera–Wadleigh SD, Badner JA, Berrettini WH et al. A high–density genome scan detects evidence for a bipolar–disorder susceptibility locus on 13q32 and other potential loci on 1q32 and 18p1 1.2. Proc Natl Acad Sci USA 1999;96(10) :5604–5609.        [ Links ]

76. Stine OC, Xu J, Koskela R et al. Evidence for linkage of bipolar disorder to chromosome 18 with a parent–of–origin effect. Am J Hum Genet 1995;57(6):1384–1394.        [ Links ]

77. Ohnishi T, Yamada K, Ohba H et al. A promoter haplotype of the inositol monophosphatase 2 gene (IMPA2) at 18p11.2 confers a possible risk for bipolar disorder by enhancing transcription. Neuropsychopharmacology 2007;32(8):1727–737.        [ Links ]

78. Sjoholt G, Ebstein RP, Lie RT et al. Examination of IMPA1 and IMPA2 genes in manic–depressive patients: association between IMPA2 promoter polymorphisms and bipolar disorder. Mol Psychiatry 2004;9(6):621–629.        [ Links ]

79. Dimitrova A, Milanova V, Krastev S et al. Association study of myo–inositol monophosphatase 2 (IMPA2) polymorphisms with bipolar affective disorder and response to lithium treatment. Pharmacogenomics J 2005;5(1):35–41.        [ Links ]

80. Kato T, Kato N. Mitochondrial dysfunction in bipolar disorder. Bipolar Disord 2000;2(3 Pt 1):180–190.        [ Links ]

81. Kato T, Kunugi H, Nanko S, Kato N. Association of bipolar disorder with the 5178 polymorphism in mitochondrial DNA. Am J Med Genet 2000;96(2):182–186.        [ Links ]

82. Washizuka S, Ikeda A, Kato N, Kato T. Possible relationship between mitochondrial DNA polymorphisms and lithium response in bipolar disorder. Int J Neuropsychopharmacol 2003;6(4) :421–424.        [ Links ]

83. Masui T, Hashimoto R, Kusumi I et al. A possible association between missense polymorphism of the breakpoint cluster region gene and lithium prophylaxis in bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry 2008;32(1):204–208.        [ Links ]

84. Serretti A, Lorenzi C, Lilli R, Mandelli L, Pirovano A et al. Pharmacogenetics of lithium prophylaxis in mood disorders: analysis of COMT, MAO–A, and Gbeta3 variants. Am J Med Genet 2002;114:370–379.        [ Links ]

85. Turecki G, Grof P, Cavazzoni P, Duffy A, Grof E et al. MAO A: Association and linkage studies with lithium responsive bipolar disorder. Psychiatric Genetics 1999;9:13–16.        [ Links ]

86. Serretti A, Lilli R, Lorenzi C, Gasperini M, Smeraldi E. Tryptophan hydroxilase gene and response to lithium prophylaxis in mood disorders. J Psychiatric Research 1999;33:371–377.        [ Links ]

87. Duffy A, Turecki G, Grof P et al. Association and linkage studies of candidate genes involved in GABAergic neurotransmission in lithium–responsive bipolar disorder. J Psychiatry Neuroscience 2000;25:353–358.        [ Links ]

88. Serretti A, Lorenzi C, Lilli R, Smeraldi E. Serotonin receptor 2A, 2C, 1A genes and response to lithium prophylaxis in mood disorders. J Psychiatric Research 2000;34:89–98.        [ Links ]

89. Alda M, Turecki G, Grof P, Cavazzoni P, Duffy A et al. Association and linkage studies of CRH and PENK genes in bipolar disorder: a collaborative IGSLI study. Am J Med Genet 2000;96:178–181.        [ Links ]

90. Szczepankiewicz A, Rybakowski J, Suwalska A et al. Association study of the glycogen synthase kinase–3 beta gene polymorphism with prophylactic lithium response in bipolar patients. World J Biological Psychiatry 2006;7(3):158–161.        [ Links ]

91. Mamdani F, Sequeira A, Alda M, Grof P, Rouleau G. No association between the PREP gene and lithium responsive bipolar disorder. BMC Psychiatry 2007;7(9). Published online 2007 February 26. doi: 10.1186/1471–244X–7–9.        [ Links ]

92. Post R, Uhde T, Roy–Byrne P, Joffe R. Correlates of antimanic response to carbamazepine. Psychiatry Research 1987;21:71–83.        [ Links ]

93. Swann A, Bowden C, Morris D, Calabrese J, Petty J et al. Depression during mania. Treatment response to lithium or valproate. Archives General Psychiatry 1997;54:37–42.        [ Links ]

94. Kruger S, Alda M, Trevor Young L, Goldapple K, Parikh S et al. Risk and resilience markers in bipolar disorder: brain responses to emotional challenge in bipolar patients and their healthy siblings. American J Psychiatry 2006;163(2):257–264.        [ Links ]

95. Nnadi C, Goldberg J, Malhotra A. Pharmacogenetics of mood disorder. Current Opinion Psychiatry 2005;18:33–39.        [ Links ]

96. Wang J, Bown C, Chen B, Trevor Young L. Identification of mood stabilizer–regulated genes by differential–display PCR. International J Neuropsychopharmacology 200;4:65–74.        [ Links ]

97. Bowden C. Predictors of response to divalproex and lithium. J Clin Psychiatry 1995;56(Supl. 3):25–30.        [ Links ]

98. Calabrese J, Rapport D, Kimmel S, Reece B, Woyshville M. Rapid cycling bipolar disorder and its treatment with valproate. Can J Psychiatry 1993;38:S57–S61.        [ Links ]

99. Calabrese J, Woyshville M, Kimmel S, Rapport D. Predictors of valproate response in bipolar rapid cycling. J Clinical Psychopharmacology 1993;13:280–283.        [ Links ]

100. Garnham J, Munro A, Slaney C et al. Prophylactic treatment response in bipolar disorder: Results of a naturalistic observation study. J Affective Disorders 2007;104:185–190.        [ Links ]

101. Reeves RR, Struve FA, Patrick G. Does EEG predict response to valproate versus lithium in patients with mania? Ann Clin Psychiatry 2001; 13 (2): 69–73.        [ Links ]

102. Greil W, Kleindienst N, Erazo N, Muller–Oerlinghausen B. Differential response to lithium and carbamazepine in the prophylaxis of bipolar disorder. J Clinical Psychopharmacology 1998;18:455–460.        [ Links ]

103. Kleindienst N, Greil W. Differential Efficacy of Lithium and Carbamazepine in the Prophylaxis of Bipolar Disorder: Results of the MAP study. Neuropsychobiology 2000;42(Supl. 1):2–10.        [ Links ]

104. Passmore J, Garnham J, Duffy A et al. Phenotypic spectra of bipolar disorder in responders to lithium versus lamotrigine. Bipolar Disorders 2003;5:110–114.        [ Links ]

105. Obrocea G, Dunn R, Frye M et al. Clinical predictors of response to lamotrigine and gabapentin monotherapy in refractory affective disorders. Biological Psychiatry 2002;51:253–260.        [ Links ]

106. Prien R, Caffey E, Klett J. Factors associated with treatment success in lithium carbonate prophylaxis. Report of the Veterans Administration and National Institute of Mental Health Collaborative Study Group. Archives General Psychiatry 1974;31:189–192.        [ Links ]

107. Steen V, Gulbrandsen A, Eiken H, Berle J. Lack of genetic variation in the coding region of the myoinositol monophosphatase gene in lithium–treated patients with manic–depressive illness. Pharmacogenetics 1996;6:113–116.        [ Links ]

108. Turecki G, Alda M, Grof P, Joober R, Cavazzoni P et al. Polyglutamine tracts: no evidence of major role in bipolar disorder. Molecular Psychiatry 1999;4:220–221.        [ Links ]

109. Turecki G, Alda M, Grof P, Joober R, Lafreniere R, Cavazzoni P. Polyglutamine coding genes in bipolar disorder: lack of association with selected candidate loci. J Affective Disorders 2000;58:63–68.        [ Links ]

110. Alda M, Grof P, Grof E. MN blood groups and bipolar disorder: evidence of genotypic association and Hardy–Weinberg disequilibrium. Biological Psychiatry 1998;44:361–363.        [ Links ]

111. Sun X, Trevor Young L, Wang J et al. Identification of lithium–regulated genes in cultured lymphoblasts of lithium–responsive subjects with bipolar disorder. Neuropsychopharmacology 2004;29:799–804.        [ Links ]

112. Turecki G, Alda M, Grof P, Martin R, Cavazzoni P et al. No association between chromosome–18 markers and lithium–responsive affective disorders. Psychiatry Research 1996;63:17–23.        [ Links ]

113. Turecki G, Grof P, Cavazzoni P et al. Lithium responsive bipolar disorder, unilineality, and chromosome 18: a linkage study. Am J Med Genet 1999;88:411–415.        [ Links ]

 

NOTE

* This paper is based on the lecture delivered by doctor Martin Alda at the XXI Research Conference of the National Institute of Psychiatry, on October 2006, and updated with recent published data.

Creative Commons License Todo el contenido de esta revista, excepto dónde está identificado, está bajo una Licencia Creative Commons