Elsevier

Brain Research Reviews

Volume 33, Issues 2–3, September 2000, Pages 228-241
Brain Research Reviews

Full-length review
Cortistatin: a member of the somatostatin neuropeptide family with distinct physiological functions

https://doi.org/10.1016/S0165-0173(00)00031-XGet rights and content

Abstract

Cortistatin is a recently discovered neuropeptide relative of somatostatin named after its predominantly cortical expression and ability to depress cortical activity. Cortistatin-14 shares 11 of the 14 amino acids of somatostatin-14 yet their nucleotide sequences and chromosomal localization clearly indicate they are products of separate genes. Now cloned from human, mouse and rat sources, cortistatin is known to bind all five cloned somatostatin receptors and share many pharmacological and functional properties with somatostatin including the depression of neuronal activity. However, cortistatin also has many properties distinct from somatostatin including induction of slow-wave sleep, apparently by antagonism of the excitatory effects of acetylcholine on the cortex, reduction of locomotor activity, and activation of cation selective currents not responsive to somatostatin. Expression of mRNA encoding cortistatin follows a circadian rhythm and is upregulated on deprivation of sleep, suggesting cortistatin is a sleep modulatory factor. This review summarizes recent advances in our understanding of the neurobiology of cortistatin, examines the similarities and differences between cortistatin and somatostatin, and asks the question: does cortistatin bind to a cortistatin-specific receptor?

Section snippets

Cloning, chromosomal mapping and structure of the cortistatin gene

The cDNA encoding cortistatin was isolated through characterization of regional specific rat brain mRNAs using directional tag PCR subtractive hybridization by de Lecea et al. in 1996 [19]. The sequence indicated the clone to be a novel 112 amino acid protein with a striking homology to somatostatin at its distal C-terminal end. The C-terminal 14 predicted amino acids, which are preceded by a pair of lysines, potentially forming a site for proteolytic cleavage, share 11 residue identities with

Cortistatin regulatory elements

Analysis of the regulatory elements for the preprocortistatin gene, recently determined by Calbet et al. [9], indicates that the regulatory elements of preprocortistatin and preprosomatostatin genes share few similarities (<30% using the BESTFIT program). The preprocortistatin transcriptional regulatory region lacks a TATA box or Initiator element and is GC rich in the proximal region. The promoter region contains a GATA motif at the −107 position which raises the possibility of basal

Regulation of cortistatin mRNA expression

The pattern of accumulation of cortistatin mRNA during development was examined using Northern blot and in situ hybridization. The 600 nucleotide band corresponding to preprocortistatin was observed in rats initially between P5 and P10, achieving maximal levels by P15, and subsequently reducing slightly in intensity into adulthood [20]. By in situ hybridization preprocortistatin mRNA was detected in the cortical plate, subiculum and stratum oriens of the hippocampus starting at P0. By P5 the

Localization of cortistatin

Using the technique of in situ hybridization, cortistatin mRNA has been shown to be essentially restricted in its expression to the cortex and hippocampus (Fig. 4A). This is in contrast to the expression of its relative somatostatin which is widely expressed in central and peripheral tissues [59], [71]. The brain specific nature of cortistatin expression was originally indicated by Northern blot: cortistatin mRNA was found in rat brain but not in adrenal gland, liver, spleen, thymus, ovary,

Structure of the cortistatin peptide

The putative 112 amino acid protein encoded by the preprocortistatin gene contains multiple sites at which endoproteolysis may occur. Recently Puebla et al. [65] have shown that preprocortistatin is indeed cleaved at the two C-terminal dibasic cleavage sites, KK and KR, to produce rCST14 and rCST29 analogously to SST14 and SST28. However, evidence of cleavage at both C-terminal dibasic sites to produce rCST14 and rCST13 was not found. The ratio of rCST14:rCST29:preprocortistatin in tissue

Cortistatin pharmacology

Given the apparent structural homologies between cortistatin and somatostatin the first target receptors tested for CST14 binding were the somatostatin receptors. Somatostatin binds to five different known receptors, named SSTR1-5, which are members of the 7-transmembrane G-protein coupled receptor superfamily. The receptors are widely distributed in the brain, and periphery, with distinct patterns of expression [38], [59], [71]. SSTRs transduce ligand-binding into cellular effects via G

Cortistatin electrophysiology

The first observation of the electrophysiological effects of cortistatin was superfusion of rCST14 on the hippocampal slice preparation [19]. The rCST14 hyperpolarized current- and voltage-clamped hippocampal neurons as evinced by inhibition of action potential firing, in a similar manner to SST14. The effect of rCST29 on the same preparation was indistinguishable from that of rCST14 (de Lecea, unpublished data). As a means to explain the mechanism of cortistatin-induced inhibition, the effect

Behavioral and neurobiological effects of cortistatin

Cortistatin was originally shown to have sleep promoting properties and effects on animal locomotor behavior. Infusion of up to 6 nmol rCST14 into rat brain ventricles induced hypoactivity in the treated rat population. The EEG for rCST14 treated animals showed a significant increase in cortical slow waves (1–4 Hz), and polygraphic recording indicated that the rCST14 injected animals spent significantly more time in slow wave sleep and less time in paradoxical (REM) sleep than control injected

Sleep promoting properties

The observation that cortistatin is mainly expressed in the cortex, and in particular, in local circuit inhibitory neurons, led to the hypothesis that the endogenous peptide may have a role in modulating cortical activity. Indeed, ICV injection of nanomolar amounts of CST14 significantly increased the number and duration of slow-wave sleep episodes both in normal and reversed light/dark cycles.

How does cortistatin promote sleep? Initial experiments have shown that application of CST14

Cortistatin and neuroprotection

Cortistatin has also been demonstrated to be neuroprotective against kainate-induced neurotoxicity in rat brains. Injection of the excitotoxic seizure inducing compound kainic acid (KA) caused seizure behavior in injected rats, which could be significantly reduced by prior ICV injection of rCST14. KA also caused significant neuronal death in the CA1 and CA3 regions of the hippocampus, an effect that could be blocked by application of rCST14 [7]. The neuroprotective properties of cortistatin are

Cortistatin in the somatostatin-like Cys–Cys loop family of peptides

Cortistatin shares many structural and functional properties with somatostatin. The amino acid sequences of the bioactive peptides, gene structures, partial coexpression, activation of common receptors and signaling pathways, and inhibition of neurons via activation of the M-current all indicate a duplication of function between these two peptides. Indeed the lack of a significant phenotype in mice lacking the gene for somatostatin [36] suggests that cortistatin at least partly duplicates

Does cortistatin have a specific receptor?

It is clear that cortistatin is capable of activating all known somatostatin receptors. However, it has not yet been shown that the observed in vitro or artificially induced activation of somatostatin receptors reflects in vivo events. Additionally, a large body of experimental evidence, discussed above, actually points towards the in vivo effects of cortistatin being mediated by receptors which are distinct from the classically defined somatostatin receptors. In summary this evidence includes:

Acknowledgements

This work was supported in part by grants from NIH (MH58543 and AG17354) to LdL. ADS is a NRSA fellow.

References (87)

  • J.C Dunlap

    Molecular bases for circadian clocks

    Cell

    (1999)
  • J.F Flood et al.

    The cortical neuropeptide, cortistatin-14, impairs post-training memory processing

    Brain Res.

    (1997)
  • S Fukusumi et al.

    Identification and characterization of a novel human cortistatin-like peptide

    Biochem. Biophys. Res. Commun.

    (1997)
  • A.S Galanopoulou et al.

    Heterologous processing of prosomatostatin in constitutive and regulated secretory pathways. Putative role of the endoproteases furin, PC1, and PC2

    J. Biol. Chem.

    (1993)
  • R Gardette et al.

    Modulation by somatostatin of glutamate sensitivity during development of mouse hypothalamic neurons in vitro

    Brain Res. Dev. Brain Res.

    (1995)
  • R.H Goodman et al.

    Somatostatin gene regulation: an overview

    Metabolism

    (1990)
  • J.V Halliwell et al.

    Voltage-clamp analysis of muscarinic excitation in hippocampal neurons

    Brain Res.

    (1982)
  • T Hashimoto et al.

    Induction of somatostatin by kainic acid in pyramidal and granule cells of the rat hippocampus

    Neurosci. Res.

    (1991)
  • G Lo Conte et al.

    Lesions of cholinergic forebrain nuclei: changes in avoidance behavior and scopolamine actions

    Pharmacol. Biochem. Behav.

    (1982)
  • A Luthi et al.

    H-current: properties of a neuronal and network pacemaker

    Neuron

    (1998)
  • A Marchese et al.

    Novel GPCRs and their endogenous ligands: expanding the boundaries of physiology and pharmacology

    Trends Physiol. Sci.

    (1999)
  • F Marrosu et al.

    Microdialysis measurement of cortical and hippocampal acetylcholine release during sleep-wake cycle in freely moving cats

    Brain Res.

    (1995)
  • J.D Molkentin et al.

    Cooperative activation of muscle gene expression by MEF2 and myogenic bHLH proteins

    Cell

    (1995)
  • J.D Molkentin et al.

    Phosphorylation of the MADS-Box transcription factor MEF2C enhances its DNA binding activity

    J. Biol. Chem.

    (1996)
  • B O’Dowd et al.

    The cloning and chromosomal mapping of two novel human opioid-somatostatin-like receptor genes, GPR7 and GPR8, expressed in discrete areas of the brain

    Genomics

    (1995)
  • Y.C Patel

    Somatostatin and its receptor family

    Front Neuroendocrinol.

    (1999)
  • J Perez et al.

    Functional effects of D-Phe-c[Cys-Tyr-D-Trp-Lys-Val-Cys]-Trp-NH2 and differential changes in somatostatin receptor messenger RNAs, binding sites and somatostatin release in kainic acid-treated rats

    Neuroscience

    (1995)
  • M Rezek et al.

    Behavioural and motor excitation and inhibition induced by the administration of small and large doses of somatostatin into the amygdala

    Neuropharmacology

    (1977)
  • B.L Roth et al.

    What’s all the RAVE about receptor internalization?

    Neuron

    (1999)
  • M Schindler et al.

    Somatostatin receptors in the central nervous system

    Prog. Neurobiol.

    (1996)
  • G Sperk et al.

    Kainic acid induced seizures: changes in somatostatin, substance P and neurotensin

    Neuroscience

    (1986)
  • D.J Stewart et al.

    Cholinergic activation of the electrocorticogram: role of the substantia innominata and effects of atropine and quinuclidinyl benzilate

    Brain Res.

    (1984)
  • A Takazawa et al.

    Cysteamine potentiates entorhinal activation of dentate gyrus granule cells in rats

    Brain Res. Bull.

    (1994)
  • L Vecsei et al.

    Comparative studies with somatostatin and cysteamine in different behavioral tests on rats

    Pharmacol. Biochem. Behav.

    (1984)
  • A Vezzani et al.

    A peptidase-resistant cyclic octapeptide analogue of somatostatin (SMS 201-995) modulates seizures induced by quinolinic and kainic acids differently in the rat hippocampus

    Neuropharmacology

    (1991)
  • F. Abel, K. Ejeskar, R.M. Sjoberg, P. Kogner, T. Martinsson, Fine mapping of a neuroblastoma tumor suppressor candidate...
  • O.M Andrisani et al.

    Identification of the promoter sequences involved in the cell specific expression of the rat somatostatin gene

    Nucl. Acids Res.

    (1987)
  • L Beranek et al.

    Central administration of the somatostatin analog octreotide induces captopril-insensitive sleep responses

    Am. J. Physiol.

    (1999)
  • L Beranek et al.

    Changes in rat sleep after single and repeated injections of the long-acting somatostatin analog octreotide

    Am. J. Physiol.

    (1997)
  • C.G Celesia et al.

    Acetylcholine released from cerebral cortex in relation to state of activation

    Neurology

    (1966)
  • J Chambers et al.

    Melanin-concentrating hormone is the cognate ligand for the orphan G-protein-coupled receptor SLC-1

    Nature

    (1999)
  • I.P Chessell et al.

    Operational characteristics of somatostatin receptors mediating inhibitory actions on rat locus coeruleus neurones

    Br. J. Pharmacol.

    (1996)
  • M Connor et al.

    Cortistatin increase of a potassium conductance in rat locus coeruleus in vitro

    Br. J. Pharmacol.

    (1997)
  • Cited by (0)

    View full text