Prepulse inhibition

Prepulse inhibition: preceding stimulus attenuates the startle response.

Prepulse Inhibition (PPI) is a neurological phenomenon in which a weaker prestimulus (prepulse) inhibits the reaction of an organism to a subsequent strong startling stimulus (pulse). The stimuli are usually acoustic, but tactile stimuli (e.g. via air puffs onto the skin) [1] and light stimuli [2] are also used.

The reduction of the amplitude of startle reflects the ability of the nervous system to temporarily adapt to a strong sensory stimulus when a preceding weaker signal is given to warn the organism. PPI is detected in numerous species ranging from mice to human. Although the extent of the adaptation affects numerous systems, the most comfortable to measure are the muscular reactions, which are normally diminished as a result of the nervous inhibition.

Deficits of prepulse inhibition manifest in the inability to filter out the unnecessary information; they have been linked to abnormalities of sensorimotor gating. Such deficits are noted in patients suffering from illnesses like schizophrenia and Alzheimer's disease, and in people under the influence of drugs, surgical manipulations, or mutations. Human studies of PPI have been summarised in reviews by Braff et al. (2001)[3] and Swerdlow et al. (2008).[4]

PPI and startle reflex apparatus for mice

Procedure

PPI measurement in human.

The main three parts of the procedure are prepulse, startle stimulus, and startle reflex. Different prepulse-to-pulse intervals, or lead intervals, are used: 30, 60, 120, 240 and 480 ms. Lead interval counts from the start of prepulse to the start of the pulse. With the interval exceeding 500 ms, prepulse facilitation - increased response - is most likely to follow.[5]

A burst of white noise is usually used as the acoustic startle stimulus. Typical durations are 20 ms for prepulse and 40 ms for pulse. Background noise with 65-70 dB is used in human studies, and 30-40 dB in rodent experiments. Prepulse is typically set 3-12 dB louder than background. Startle response is measured in rodents using the so-called automated "startle chambers" or "stabilimeter chambers", with detectors recording whole-body reaction.[5]

In humans, the movements of oculomotor muscles ("eye-blink reflex" or "eye-blink response" assessed using electromyographic recording of orbicularis oculi muscle and by oculography) could be used as a measure. Pulse-alone results are compared to prepulse-plus-pulse, and the percentage of the reduction in the startle reflex represents prepulse inhibition. Possible hearing impairment must be taken into account, as, for example, several strains of mice develop high frequency hearing loss when they mature.[5]

Major features

History of Scientific Work

Prepulse inhibition was first described in 1862 by Sechenov and was rediscovered at least twice before Howard S. Hoffman discovered it again in 1963. Hoffman was the first to use the term prepulse. He continued his work on startle into the 1980s. His many papers on the reflex and its modification laid the groundwork for the widespread use of prepulse inhibition today in studies of schizophrenia and other disorders.[16]

One possible reason it was discovered and rediscovered is that it got confused with Pavlovian conditioning. Another is that it didn't fit nicely into theories of the times.[17]

Disruption of PPI

Disruptions of PPI are studied in humans and many other species. The most studied are deficits of PPI in schizophrenia, although this disease is not the only one to be associated with such deficits. They have been noted in panic disorder (Ludewig, et al., 2005), schizotypal personality disorder,[18] obsessive-compulsive disorder(Swerdlow et al., 1993), Huntington's disease,[19] nocturnal enuresis and attention deficit disorder (Ornitz et al. 1992), and Tourette's syndrome (Swerdlow et al. 1994; Castellanos et al. 1996). According to one study, people who have temporal lobe epilepsy with psychosis also show decreases in PPI, unlike those who have TLE without psychosis.[20] Therefore, PPI deficits are not typical to specific disease, but rather tell of disruptions in a specific brain circuit.

PPI deficit in schizophrenia

PPI deficits represent a well-described finding in schizophrenia, with the first report dating back to 1978.[21] The abnormalities are also noted in unaffected relatives of the patients.[22][23] In one study, patients failed to show increased PPI to attended prepulses.[24] Dopamine, which plays a major role in schizophrenia, had been shown to regulate sensorimotor gating in rodent models.[25][26] These findings fit to the dopamine hypothesis of schizophrenia. In theory, PPI disruption in schizophrenia may be related to the processes of sensory flooding and cognitive fragmentation.

Antipsychotic medication have been shown to increase PPI in patients, with atypical antipsychotics having more effect. Patients display the same gender difference in PPI as healthy people: males have higher PPI compared to females. One notable finding is that patients are specifically deficient in PPI with 60 ms prepulse intervals relative to intervals of other lengths; this remains so even under antipsychotic treatment.[27]

The other fact is the influence of cigarette smoking. Non-smoking patients have lower PPI compared to smokers, and heavy smokers have the highest PPI.[27][28] This finding runs in accord with the high rates of smoking among schizophrenic patients, estimated at 70%,[29] with many patients smoking more than 30 cigarettes a day.[30] Some studies show association of schizophrenia with the CHRNA7 and CHRFAM7A genes, which code for alpha7 subunit of nicotinic receptors, but other studies are negative.[31][32] Contrary to the predictions, nicotine receptor alpha7 subunit knockout mice do not show disruptions in PPI.[33]

Disruption of PPI in rodents

Murine models are widely used to test hypotheses linking genetic components of various diseases with sensorimotor gating. While some of the hypotheses stand to the test, others are not, as some mice models show unchanged or increased PPI contrary to the expectations, as in the tests of COMT-deficient mice.[34]

Certain surgical procedures also disrupt PPI in animals, helping to unravel the underlying circuitry.

Many animal studies of PPI are undertaken in order to understand and model the pathology of schizophrenia.[35] Schizophrenia-like PPI disruption techniques in rodents have been classified in one review[36] into four models:

Diverse chemical compounds are tested on animals with such deficits. Compounds that are able to restore PPI could be further investigated for their potential antipsychotic role.

An updated summary of both preclinical and clinical findings with PPI can be found in a recent comprehensive review.[4]

See also

References

  1. Braff, DL; Grillon, C; Geyer, MA (1992). "Gating and habituation of the startle reflex in schizophrenic patients". Archives of General Psychiatry. 49 (3): 206–15. doi:10.1001/archpsyc.1992.01820030038005. PMID 1567275.
  2. Weber, M; Swerdlow, NR (2008). "Rat strain differences in startle gating-disruptive effects of apomorphine occur with both acoustic and visual prepulses". Pharmacology, Biochemistry, and Behavior. 88 (3): 306–11. doi:10.1016/j.pbb.2007.08.014. PMC 2266874Freely accessible. PMID 17900675.
  3. Braff, DL; Geyer, MA; Swerdlow, NR (2001). "Human studies of prepulse inhibition of startle: Normal subjects, patient groups, and pharmacological studies". Psychopharmacology. 156 (2–3): 234–58. doi:10.1007/s002130100810. PMID 11549226.
  4. 1 2 Swerdlow, NR; Weber, M; Qu, Y; Light, GA; Braff, DL (2008). "Realistic expectations of prepulse inhibition in translational models for schizophrenia research". Psychopharmacology. 199 (3): 331–88. doi:10.1007/s00213-008-1072-4. PMC 2771731Freely accessible. PMID 18568339.
  5. 1 2 3 Geyer, MA; McIlwain, KL; Paylor, R (2002). "Mouse genetic models for prepulse inhibition: An early review". Molecular Psychiatry. 7 (10): 1039–53. doi:10.1038/sj.mp.4001159. PMID 12476318.
  6. Graham, FK (1975). "Presidential Address, 1974. The more or less startling effects of weak prestimulation". Psychophysiology. 12 (3): 238–48. doi:10.1111/j.1469-8986.1975.tb01284.x. PMID 1153628.
  7. Swerdlow, NR; Geyer, MA; Braff, DL (2001). "Neural circuit regulation of prepulse inhibition of startle in the rat: Current knowledge and future challenges". Psychopharmacology. 156 (2–3): 194–215. doi:10.1007/s002130100799. PMID 11549223.
  8. Paylor, R.; Crawley, Jacqueline N. (1997). "Inbred strain differences in prepulse inhibition of the mouse startle response". Psychopharmacology. 132 (2): 169–80. doi:10.1007/s002130050333. PMID 9266614.
  9. Aasen, I; Kolli, L; Kumari, V (2005). "Sex effects in prepulse inhibition and facilitation of the acoustic startle response: Implications for pharmacological and treatment studies". Journal of psychopharmacology (Oxford, England). 19 (1): 39–45. doi:10.1177/0269881105048890. PMID 15671127.
  10. Hoffman, HS; Stitt, CL (1980). "Inhibition of the glabella reflex by monaural and binaural stimulation". Journal of experimental psychology. Human perception and performance. 6 (4): 769–76. doi:10.1037/0096-1523.6.4.769. PMID 6449543.
  11. Kumari, V; Fannon, D; Sumich, AL; Sharma, T (2007). "Startle gating in antipsychotic-naïve first episode schizophrenia patients: One ear is better than two". Psychiatry Research. 151 (1–2): 21–8. doi:10.1016/j.psychres.2006.09.013. PMID 17382404.
  12. Amsterdam; New York : Elsevier, 2001 Attraction, Distraction and Action: multiple perspectives on attentional capture; By Charles L. Folk, Bradley S. Gibson. ISBN 0-444-50676-4 Google books
  13. Wu, MF (1984). "Startle reflex inhibition in the rat: its persistence after extended repetition of the inhibitory stimulus". Journal of Experimental Psychology: Animal Behavior Processes. 10 (2): 221–228. doi:10.1037/0097-7403.10.2.221.
  14. Filion, DL; Dawson, ME; Schell, AM (1993). "Modification of the acoustic startle-reflex eyeblink: A tool for investigating early and late attentional processes". Biological Psychology. 35 (3): 185–200. doi:10.1016/0301-0511(93)90001-O. PMID 8218613.
  15. Hoffman, HS; Fleshler, M (1963). "Startle Reaction: Modification by Background Acoustic Stimulation". Science. 141 (3584): 928–30. Bibcode:1963Sci...141..928H. doi:10.1126/science.141.3584.928. PMID 14043340.
  16. Obituary: Howard S. Hoffman (1925-2006), APA PsycNET American Psychological Association
  17. Ison, JR; Hoffman, HS (1983). "Reflex modification in the domain of start; II. The anomalous history of a robust and ubiquitous phenomenon". Psychological Bulletin. 94 (1): 3–17. doi:10.1037/0033-2909.94.1.3. PMID 6353464.
  18. Cadenhead, KS; Geyer, MA; Braff, DL (1993). "Impaired startle prepulse inhibition and habituation in patients with schizotypal personality disorder". The American Journal of Psychiatry. 150 (12): 1862–7. PMID 8238643.
  19. Swerdlow, NR; Paulsen, J; Braff, DL; Butters, N; Geyer, MA; Swenson, MR (1995). "Impaired prepulse inhibition of acoustic and tactile startle response in patients with Huntington's disease". Journal of neurology, neurosurgery, and psychiatry. 58 (2): 192–200. doi:10.1136/jnnp.58.2.192. PMC 1073317Freely accessible. PMID 7876851.
  20. Morton, N., Gray, N.S., Mellers, J., Toone, B., Lishman, W.A., & Gray, J.A. (1994). Prepulse inhibition in temporal lobe epilepsy. Schizophrenic Research, 15, 191.
  21. Braff, D; Stone, C; Callaway, E; Geyer, M; Glick, I; Bali, L (1978). "Prestimulus effects on human startle reflex in normals and schizophrenics". Psychophysiology. 15 (4): 339–43. doi:10.1111/j.1469-8986.1978.tb01390.x. PMID 693742.
  22. Kumari, V; Das, M; Zachariah, E; Ettinger, U; Sharma, T (2005). "Reduced prepulse inhibition in unaffected siblings of schizophrenia patients". Psychophysiology. 42 (5): 588–94. doi:10.1111/j.0048-5772.2005.00346.x (inactive 2015-02-01). PMID 16176381.
  23. Cadenhead, KS; Swerdlow, NR; Shafer, KM; Diaz, M; Braff, DL (2000). "Modulation of the startle response and startle laterality in relatives of schizophrenic patients and in subjects with schizotypal personality disorder: Evidence of inhibitory deficits". The American Journal of Psychiatry. 157 (10): 1660–8. doi:10.1176/appi.ajp.157.10.1660. PMID 11007721.
  24. Hazlett, EA; Romero, MJ; Haznedar, MM; New, AS; Goldstein, KE; Newmark, RE; Siever, LJ; Buchsbaum, MS (2007). "Deficient attentional modulation of startle eyeblink is associated with symptom severity in the schizophrenia spectrum". Schizophrenia Research. 93 (1–3): 288–95. doi:10.1016/j.schres.2007.03.012. PMID 17478083.
  25. Mansbach, RS; Geyer, MA; Braff, DL (1988). "Dopaminergic stimulation disrupts sensorimotor gating in the rat". Psychopharmacology. 94 (4): 507–14. doi:10.1007/BF00212846. PMID 3131796.
  26. Swerdlow, NR; Keith, VA; Braff, DL; Geyer, MA (1991). "Effects of spiperone, raclopride, SCH 23390 and clozapine on apomorphine inhibition of sensorimotor gating of the startle response in the rat". The Journal of Pharmacology and Experimental Therapeutics. 256 (2): 530–6. PMID 1825226.
  27. 1 2 Swerdlow, NR; Light, GA; Cadenhead, KS; Sprock, J; Hsieh, MH; Braff, DL (2006). "Startle gating deficits in a large cohort of patients with schizophrenia: Relationship to medications, symptoms, neurocognition, and level of function". Archives of General Psychiatry. 63 (12): 1325–35. doi:10.1001/archpsyc.63.12.1325. PMID 17146007.
  28. Kumari, V; Soni, W; Sharma, T (2001). "Influence of cigarette smoking on prepulse inhibition of the acoustic startle response in schizophrenia". Human psychopharmacology. 16 (4): 321–326. doi:10.1002/hup.286. PMID 12404567.
  29. Leonard, S; Adler, LE; Benhammou, K; Berger, R; Breese, CR; Drebing, C; Gault, J; Lee, MJ; et al. (2001). "Smoking and mental illness". Pharmacology, Biochemistry, and Behavior. 70 (4): 561–70. doi:10.1016/S0091-3057(01)00677-3. PMID 11796154.
  30. De Leon, J; Tracy, J; McCann, E; McGrory, A; Diaz, FJ (2002). "Schizophrenia and tobacco smoking: A replication study in another US psychiatric hospital". Schizophrenia Research. 56 (1–2): 55–65. doi:10.1016/S0920-9964(01)00192-X. PMID 12084420.
  31. Gene Overview of All Published Schizophrenia-Association Studies for CHRFAM7A – Schizophrenia Gene Database.
  32. Gene Overview of All Published Schizophrenia-Association Studies for CHRNA7 – Schizophrenia Gene Database
  33. Paylor, R; Nguyen, M; Crawley, JN; Patrick, J; Beaudet, A; Orr-Urtreger, A (1998). "Alpha7 nicotinic receptor subunits are not necessary for hippocampal-dependent learning or sensorimotor gating: A behavioral characterization of Acra7-deficient mice". Learning & memory (Cold Spring Harbor, N.Y.). 5 (4–5): 302–16. PMC 311270Freely accessible. PMID 10454356.
  34. Gogos, JA; Morgan, M; Luine, V; Santha, M; Ogawa, S; Pfaff, D; Karayiorgou, M (1998). "Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior". Proceedings of the National Academy of Sciences of the United States of America. 95 (17): 9991–6. Bibcode:1998PNAS...95.9991G. doi:10.1073/pnas.95.17.9991. PMC 21449Freely accessible. PMID 9707588.
  35. Swerdlow, NR; Geyer, MA (1998). "Using an animal model of deficient sensorimotor gating to study the pathophysiology and new treatments of schizophrenia". Schizophrenia bulletin. 24 (2): 285–301. doi:10.1093/oxfordjournals.schbul.a033326. PMID 9613626.
  36. Geyer, MA; Krebs-Thomson, K; Braff, DL; Swerdlow, NR (2001). "Pharmacological studies of prepulse inhibition models of sensorimotor gating deficits in schizophrenia: A decade in review". Psychopharmacology. 156 (2–3): 117–54. doi:10.1007/s002130100811. PMID 11549216.

External links

This article is issued from Wikipedia - version of the 10/31/2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.