The Effect of the Trpv1 Agonist, Capsaicin, on the Developing Rat Brain: A Mini Review

Loris A Chahl

School of Biomedical Sciences and Pharmacy University of Newcastle NSW 2308 Australia


It has been shown previously that rats treated as neonates with capsaicin exhibited hyperactivity in a novel environment and had brain changes, including reduced brain weight, reduced hippocampal area, reduced cortical thickness and increased neuronal density in several cortical areas. These brain changes resembled those found in subjects with schizophrenia. This mini review discusses recent findings on the effects of capsaicin on hippocampal neurons and the role of TRPV1 channels in the central nervous system, which provide a possible explanation for the effects of capsaicin on the developing rat brain. Future studies on the role of TRPV1 channels in the brain hold great promise for further understanding of neuropsychiatric disorders.


More than 40 years ago Jancsó et al1,2 showed that treatment of neonatal rats with the vanilloid, capsaicin, the hot principle in chillies, produced life-long elimination of chemo sensitive primary afferent neurons and loss of chemogenic pain and neurogenic inflammation. Adult animals were less affected by capsaicin, exhibiting only a temporary loss of pain and inflammation. These observations followed on from the historic work of N Jancó who showed the desensitizing effect of capsaicin applied to sensory neurons3. At that time there was little understanding of the complexity of chemical neurotransmission. The concept of co-transmission was only beginning to evolve4, and the role of neuropeptides was yet to be explored.

A few years earlier Chang and Leeman5 had isolated a peptide from bovine hypothalamus and characterized it as substance P, a previously unidentified substance that had been discovered many years earlier by von Euler and Gaddum6. The discovery of the structure of substance P7 opened an era of intensive investigation of its localization and role in the nervous system8. Over the next few years substance P was demonstrated to be responsible for antidromic vasodilatation and neurogenic inflammation9 and to be released from the central synapses of small diameter primary afferent neurons on noxious stimulation10.

During the early 1980s capsaicin treatment of neonatal rats was found to produce not only permanent loss of many unmyelinated primary afferent neurons, but also depletion of substance P in regions of termination of primary afferents in the spinal cord and medulla. However, substance P in higher central nervous system (CNS) regions appeared not to be affected11. Thus, the effect of capsaicin on the nervous system was considered to be limited mainly to action on primary afferent neurons.

The discovery of the transient receptor potential vanilloid type 1 (TRPV1) channel, a heat activated, non-selective cation channel, as the site of action of capsaicin, was a major advance in the field12,13. The finding that responses to noxious heat and inflammatory pain were severely impaired in mice lacking the capsaicin receptor (then known as VR1)14 led to great interest in the development of analgesic agents which acted on this new target15-17.

The predominately primary afferent location of the site of action of capsaicin at first seemed to be confirmed when several studies showed little or very restricted localization of TRPV1 channels in the CNS12,18,19. Further studies using methods with increased sensitivity, however, showed that TRPV1 channels were not restricted to primary afferent neurons but were also widely distributed in the CNS, albeit at levels 20-30 times lower than those in the peripheral nervous system20. Thus, investigation of the role of TRPV1 channels and of the effects of capsaicin in the brain lagged behind studies of the peripheral actions.

The observations that subjects with schizophrenia have deficits in pain sensation and flare responses to niacin prompted a study by Newson et al21 of the effect of neonatal capsaicin treatment on rat brain and behaviour. At 5-7 weeks the rats treated as neonates with capsaicin had increased locomotor activity in a novel environment. The most striking finding, however, was that although they had normal body weight, the rats had reduced brain weight, reduced hippocampal and coronal cross-sectional areas, reduced cortical thickness and increased neuronal density in several cortical areas21. Some of these changes were found to persist into adulthood22.

The resemblance between the brain changes found in rats treated as neonates with capsaicin, and those found in post-mortem human brains of subjects with schizophrenia23, was considered noteworthy. A subsequent investigation by Petrovszski et al24 of the behavioural changes in young rats treated with capsaicin showed that the animals had reduced memory function, as well as disturbances in thermoregulation and motor activity. These findings raised the possibility that rats treated during development with capsaicin might be an animal model of schizophrenia. However, sensory-motor gating was not affected in either study22,24, indicating that in this regard the capsaicin treated rat did not exhibit one of the generally accepted characteristics of an animal model of schizophrenia.

A major question that arises regarding the brain changes observed in rats treated as neonates with capsaicin is whether they were due to a central or peripheral action of capsaicin. The lack of a clear demonstration of the presence of TRPV1 channels in the CNS led Newson et al21 to propose that the brain and behavioural changes seen in capsaicin treated rats were an indirect effect of the action of capsaicin on TRPV1 channels on primary afferent neurons, resulting in loss of sensory input to the brain, decreased synaptic density, and increased neuronal density. The primary afferent neuron would undoubtedly have been a significant site of action of capsaicin in neonatal rats and the importance of primary afferent input to brain development should not be dismissed. Indeed, a study by Orefice et al25 has shown that peripheral mechanosensory neuron dysfunction induced by deletion during development of Mecp2 or Gabrb3, genes associated with autism spectrum disorder in humans, resulted in social interaction deficits and anxiety-like behaviour in mice. Nevertheless, in light of more recent evidence of the presence of functional TRPV1 channels in the brain20, it is considered most likely that neonatal capsaicin treatment produced brain changes by a direct action on central TRPV1 channels.

Of particular relevance to the effects of neonatal capsaicin treatment on rat brain development are studies on the role of TRPV1 channels in neuronal growth and development26. The first studies that linked TRPV1 channels with neuronal development showed that TRPV1 channels were localised in neurites and growth cones where they were involved in the formation of filopodia in neurons27. Stock et al28 showed that TRPV1 channels are expressed in mouse neural precursor cells during postnatal development but not in the adult. However, TRPV1 channel expression was induced in the adult by activities involving spatial learning and exercise. Furthermore, loss of TRPV1 channel expression in Trpv1-deficient (Trpv1-/-) mice led to an increase in proliferation of neural precursor cells28.

A recent study by Wang et al29 has shown that Trpv1-/- mice were more stress resilient than control animals. Furthermore, glucocorticoid receptor-mediated expression and activity of the chromatin-remodeling factor, histone deacetylase 2 (HDAC2), were reduced in Trpv1-/- mice29. HDAC2 has been shown to repress gene expression of cell-cycle-related genes involved in synaptic plasticity and neuronal differentiation. Thus, reduction in HDAC2 activity facilitated hippocampal neurogenesis30. Indeed, Wang et al29 have suggested that HDAC2 is a molecular link between TRPV1 activity and stress responses. In a further investigation, capsaicin was shown to reduce synaptic density in cultured primary hippocampal neurons, and to impair the proliferation, maturation and survival of neurons in the hippocampus of adult mice. These effects of capsaicin were mediated by HDAC2, which was elevated by capsaicin31. The effects of capsaicin were absent in Trpv1-/- mice, indicating that they were mediated by TRPV1 channels.

One of the important characteristics of the action of capsaicin on primary afferent neurons is that the activation of TRPV1 channels is followed by a period of desensitization during which the cell is unresponsive to TRPV1 agonists32. Furthermore, administration of high doses of TRPV1 agonists leads to calcium overload and death of TRPV1 bearing neurons33. Recent studies have shown that supra-spinal neurons also exhibit desensitization to TRPV1 agonists34. Therefore, the resultant effects of capsaicin on the plasticity of particular neurons is likely to be a balance between the agonist action, desensitization and cell death. Treatment of neonatal rats with capsaicin would be expected to produce agonist action followed by prolonged desensitization and cell death of some TRPV1 bearing neurons. It is considered that cell death alone would be unlikely to explain the brain changes in rats treated as neonates with capsaicin21, since neuronal density was increased in several cortical areas, indicating that synaptic density rather than cell number was reduced.

Although hippocampal neuronal density was not investigated in the study by Newson et al21, in light of the findings of Wang et al31, it is proposed that the brain changes in rats treated as neonates with capsaicin21, were produced by prolonged TRPV1 agonist activity by capsaicin resulting directly or indirectly in prolonged activation of cellular pathways that lead to reduced neuronal arborisation. The finding that TRPV1 channels were expressed at greater densities in the early developmental period19,28 might explain the prolonged agonist action of capsaicin in neonatal animals and thus their greater vulnerability.

The possible role of TRPV1 channels in the central nervous system and in neuropsychiatric disorders has been increasingly studied over the past fifteen years35-39. Marsch et al40 showed that TRPV1 knock-out mice exhibited less fear, anxiety and hippocampal long- term potentiation (LTP) than wild type mice. In agreement with this study was the finding that acute administration of a TRPV1 agonist improved cognitive performance of rats in passive avoidance learning tasks41. TRPV1 channel activation has also been found to trigger another form of synaptic plasticity, interneuron long-term depression (LTD)42. Both LTP and LTD are considered to be involved in the formation of stable memories. The TRPV1 agonists, capsaicin and resiniferatoxin, facilitated LTP and suppressed LTD in rat hippocampal slices, and exerted a protective effect against the effects of stress on synaptic plasticity and spatial memory formation43. Activation of TRPV1 channels by capsaicin has also been found to decrease memory impairment in a rat model of biliary cirrhosis44, whereas a TRPV1 antagonist was found to impair discrimination between novel and familiar objects in rats45.

The role of TPVPV1 channels in neurogenesis, synaptic plasticity, learning and memory requires further investigation. Although Marsch et al40 found reduced LTP in TRPV1 knock- out mice, other investigators have reported increased LTP in TRPV1 knock-out mice46.

Moreover, the finding by Stock et al28 of increased neurogenesis in TRPV1 knock-out mice would be expected to result in increased synaptic plasticity, enhanced LTP and enhanced learning and memory, which would seem to be at odds with studies showing that knock-out mice have reduced LTP40. However, the discrepancy might be explained by the finding of less differentiation of neural precursor cells into neurons and glia in TRPV1 knock-out animals33.

The revelation that anandamide, an endogenous cannabinoid, is a TRPV1 agonist47, provided a further dimension and added complexity to studies on vanilloids in health and disease. In the hippocampal dentate gyrus, TRPV1 channel activation by endogenous anandamide has been found to trigger postsynaptic LTD46. Cross-talk between the endocannabinoid and endovanilloid receptor systems, whereby the cannabinoid CB1 receptor sensitizes TRPV1 receptors to anandamide, has been demonstrated in primary afferent neurons48. A balance or interplay between the endocannabinoid and endovanilloid systems at central synapses involved in learning, memory and cognition has been found in several studies49,50.

Furthermore, some of the effects of cannabinoids in rats, including apathetic and impulsive patterns of behaviour, have been shown to be partially mediated by TRPV1 channels51. The complex interactions between the endocannabinoid and endovanilloid systems at central synapses involved in learning, memory and cognition require further elucidation and should prove of considerable importance in advancing understanding of neuropsychiatric disorders36.

The discovery of the TRPV1 channel promised new horizons in pain control15-17 and advances in treatment of several other disorders52,53. However, the multifunctional nature of the TRPV1 channel also poses considerable challenges for targeted drug design. Recent studies on the molecular mechanism of activation of the TRPV1 channel by capsaicin54,55 may provide clues for new approaches to drug design.

Although investigation of the roles of vanilloids and cannabinoids in brain function is at an early stage, it holds the promise of greater understanding of the responses of the central nervous system to sensory input and stressors. The exploitation of such understanding to provide therapeutic advances for currently poorly understood brain and neuropsychiatric disorders remains a tantalizing possibility.

The author declares no conflict of interest.

  1. Jancsó G, Kiraly E, Jancsó-Gábor A. Pharmacologically induced selective degeneration of chemosensitive primary afferent neurons. Nature. 1977; 270: 741-743.
  2. Szolcsányi J. Forty years in capsaicin research for sensory pharmacology and physiology. Neuropeptides. 2004; 38: 377-384.
  3. Jancsó N, Jancsó-Gábor A, Szolcsányi J. Direct evidence for neurogenic inflammation and its prevention by denervation and by pretreatment with capsaicin. Br J Pharmac Chemother. 1967; 31: 138-151.
  4. Campbell G. Cotransmission. Ann Rev Pharmac Toxicol. 1987; 27: 51-70.
  5. Chang MM, Leeman SE. Isolation of a sialogogic peptide from bovine hypothalamic tissue and its characterization as substance P. J Biol Chem. 1970; 245: 4784-4790.
  6. von Euler US, Gaddum JH. An unidentified depressor substance in certain tissue extracts.J Physiol. 1931; 72: 74-87.
  7. Chang MM, Leeman SE, Niall HD. Amino-acid sequence of substance P. Nature New Biol. 1971; 232: 86-87.
  8. Chahl LA. Antidromic vasodilatation and neurogenic inflammation. Pharmac Ther. 1988; 37: 275-300.
  9. Lembeck F, Holzer P. Substance P as neurogenic mediator of antidromic vasodilation and neurogenic plasma extravasation. N S Arch Pharmacol. 1979; 310: 175-183.
  10. Duggan AW, Hendry IA. Laminar localization of the sites of release of immunoreactive substance P in the dorsal horn with antibody-coated microelectrodes. Neurosci Lett. 1986; 68: 134-140.
  11. Helke CJ, DiMicco JA, Jacobowitz DM, Kopin IJ. Effect of capsaicin administration to neonatal rats on the substance P content of discrete CNS regions. Brain Res. 1981; 222: 428-431.
  12. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997; 389: 816-824.
  13. Szallasi A and Blumberg PM. Vanilloid (capsaicin) receptors and mechanisms. Pharmac Rev. 1999; 51: 159-212.
  14. Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg M, Basbaum AI, Julius D. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science. 2000; 288: 306-313.
  15. Szallasi A, Cruz F, Geppetti P. TRV1: a therapeutic target for novel analgesic drugs? Trends Mol Med. 2006; 12: 545-554.
  16. Wong GY, Gavva NR. Therapeutic potential of vanilloid receptor TRPV1 agonists and antagonists as analgesics: Recent advances and setbacks. Brain Res Rev. 2009; 60: 267- 277.
  17. Szolcsányi J, Sándor Z. Multisteric TRPV1 nocisensor: a target for analgesics. Trends Pharmacol Sci. 2012; 33: 646-655.
  18. Szallasi A, Nilsson S, Farkas-Szallasi T, Blumberg PM, Hὅkfelt T, Lundberg IM. Vanilloid (capsaicin) receptors in the rat: distribution in the brain, regional differences in the spinal cord, axonal transport to the periphery, and depletion by systemic vanilloid treatment. Brain Res. 1995; 703: 175-183.
  19. Cavanaugh DJ, Chesler AT, Jackson AC, Sigal YM, Yamanaka H, Grant R, O’Donnell D, Nicoll RA, Shah NM, Julius D, Basbaum AI. Trpv1 reporter mice reveal highly restricted brain distribution and functional expression in arteriolar smooth muscle cells. J Neurosci. 2011; 31: 5067-5077.
  20. Martins D, Tavares I, Morgado C. “Hotheaded”: The role of TRPV1 in brain functions. Neuropharmacology. 2014; 85: 151-157.
  21. Newson P, Lynch-Frame A, Roach R, Bennett S, Carr V, Chahl LA. Intrinsic sensory deprivation induced by neonatal capsaicin treatment induces changes in rat brain and behaviour of possible relevance to schizophrenia. Brit J Pharmacol. 2005; 146: 408-418.
  22. Newson PN, van den Buuse M, Martin S, Lynch-Frame A, Chahl LA. Effects of neonatal treatment with the TRPV1 agonist, capsaicin, on adult rat brain and behaviour. Behav Brain Res. 2014; 272: 55-65.
  23. Selemon LD and Goldman-Rakic PS. The reduced neupil hypothesis: a circuit based model of schizophrenia. Biol Psychiatry. 1999; 45: 17-25.
  24. Petrovszki Z, Adam G, Kekesi G, Tuboly G, Morvay Z, Nagy E, Benedek G, Horvath G. The effect of juvenile capsaicin desensitization in rats: Behavioral impairments. Physiol Behav. 2014; 125: 38-44.
  25. Orefice LL, Zimmerman AL, Chirila AM, Sleboda SJ, Head JP, Ginty DD. Peripheral mechanosensory neuron dysfunction underlies tactile and behavioral deficits in mouse models of ASDs. Cell. 2016; 166: 299-313.
  26. Ramirez-Barrantes R, Cordova C, Poblete H, Munoz P, Marchant I, Wianny F, Olivero P. Perspectives of TRPV1 function on the neurogenesis and neural plasticity. Neural Plasticity. 2016; doi.org/10.1155/2016/1568145.
  27. Goswami C, Hucho T. TRPV1 expression-dependent initiation and regulation of filopodia. J Neurochem. 2007; 103: 1319-1333.
  28. Stock K, Garthe A, de Almeida Sassi F, Glass R, Wolf SA, Kettenmann H. The capsaicin receptor TRPV1 as a novel modulator of neural precursor cell proliferation. Stem Cells. 2014; 32:3183-3195.
  29. Wang SE, Ko SY, Jo S, Choi M, Lee SH, Jo H-R, Seo JY, Lee SH, Kim Y-S, Jung S-J, Son H. TRPV1 regulates stress responses through HDAC2. Cell Reports. 2017; 19: 401-412.
  30. Guan JS, Haggarty SJ, Giacometti E, Dannenberg JH, Joseph N, Gao J, Nieland TJ, Zhou Y, Wang X, Mazitschek R, Bradner JE, DePinho RA, Jaenisch R, Tsai L-H. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature. 2009; 459: 55–60.
  31. Wang SE, Ko SY, Kim Y-S, Jo S, Lee SH, Jung SJ, Son H. Capsaicin upregulates HDAC2 via TRPV1 and impairs neuronal maturation in mice. Exp Mol Med. 2018; 50: e455, doi:10.1038/emm.2017.289.
  32. Sanz-Salvador L, Andres-Borderia A, Ferrer-Montiel A, Planells-Cases R. Agonist- and Ca2+- dependent desensitization of TRPV1 channel targets the receptor to lysosomes for degradation. J Biol Chem. 2012; 287: 19462-19471.
  33. Olah Z, Szabo T, Karai L, Hough C, Fields RD, Caudle RM, Blumberg PM, Iadarola MJ. Ligand-induced dynamic membrane changes and cell deletion conferred by vanilloid receptor 1. J Biol Chem. 2001; 276: 11021-11030.
  34. Fukushima A, Mamada K, Iimura A, Ono H. Supraspinal-selective TRPV1 desensitization induced by intracerebroventricular treatment with resiniferatoxin. Scientific Reports. 2017; 7: 12452 doi: 10.1038/s41598-017-12717-5.
  35. Chahl LA (2011). TRP channels and psychiatric disorders. In: Transient Receptor Potential Channels. Ed MS Islam, Adv Exp Med Biol 704 Springer BV. 987-1009; 2011.
  36. Ruggiero RN, Rossignoli MT, De Ross JB, Hallak JEC, Leite JP, Bueno-Junior LS. Cannabinoids and vanilloids in schizophrenia: neurophysiological evidence and directions for basic research. Front Pharmacol. 2017; 8: 399. doi: 10.3389/fphar.2017.00399.
  37. Jayant S, Sharma BM, Sharma B. Protective effect of transient receptor potential vanilloid subtype 1 (TRPV1) modulator, against behavioral, biochemical and structural damage in experimental models of Alzheimer’s disease. Brain Res. 2016; 1642: 397-408.
  38. Tian Y-H, Lee S-Y, Kim H-C, Jang C-G. Repeated methamphetamine treatment increases expression of TRPV1 mRNA in the frontal cortex but not in the striatum or hippocampus of mice. Neurosci Letts. 2010; 472: 61-64.
  39. Jeng JH, Lee DK, Liu S-M, Chua SC, Schwartz GJ, Jo Y-H. Activation of temperature- sensitive TRPV1-like receptors in ARC POMC neurons reduces food intake. PloS Biol. 2018/16: e2004399. doi.org/10.1371/journal.pbio.2004399.
  40. Marsch R, Foeller E, Rammes G, Bunck M, Kossl M, Holsboer F, Zieglgansberger W, Landgraf R, Lutz B, Wotjak CT. Reduced anxiety, conditioned fear, and hippocampal long- term potentiation in transient receptor potential vanilloid type 1 receptor-deficient mice. J Neurosci. 2007; 27: 832-839.
  41. Shiri M, Komaki A, Oryan S, Taheri M, Komaki H, Etaee F. Effects of cannabinoid and vanilloid receptor agonists and their interaction on learning and memory in rats. Canad J Physiol Pharmacol. 2017; 95: 382-387.
  42. Gibson HE, Edwards JG, Page RS, Van Hook MJ, Kauer JA. TRPV1 channels mediate long- term depression at synapses on hippocampal interneurons. Neuron. 2008; 57: 746-759.
  43. Li H-B, Mao R-R, Zhang J-C, Yang Y, Cao J, Xu L Antistress effect of TRPV1 channel on synaptic plasticity and spatial memory. Biol Psychiatry. 2008; 64: 286-292.
  44. Bashiri H, Hosseini-Chegeni H, Sharifi KA, Sahebgharani M, Salari A-A. Activation of TRPV1 receptors affects memory function and hippocampal TRPV1 and CREB mRNA expression in a rat model of biliary cirrhosis. Neurological Res. 2018; 40: 938-947.
  45. Bannazadeh M, Fatehi F, Fatemi I, Roohbakhsh A, Allahtavakoli M, Nasiri M, Azin M, Shamsizadeh A. The role of transient receptor potential vanilloid type 1 in unimodal and multimodal object recognition task in rats. Pharmacol Reports. 2017; 69: 526-531.
  46. Chavez AE, Chiu CQ, Castillo PE. TRPV1 activation by endogenous anandamide triggers postsynaptic LTD in dentate gyrus. Nat Neurosci. 2010; 13: 1511-1518.
  47. Ross, RA. Anandamide and vanilloid TRPV1 receptors. Br J Pharmacol. 2003; 140: 790- 801.
  48. Chen J, Varga A, Selvarajah S, Jenes A, Dienes B, Sousa-Valente J, Kulik A, Veress G, Brain SD, Baker D, Urban L, Mackie K, Nagy I. Spatial distribution of the cannabinoid type 1 and capsaicin receptors may contribute to the complexity of their crosstalk. Sci Rep. 2016; 6: 33307-33325.
  49. Back FP, Carobrez AP. Periaqueductal gray glutamatergic, cannabinoid and vanilloid receptor interplay in defensive behavior and aversive memory formation. Neuropharmacology. 2018; 135: 399-411.
  50. Cui Y, Perez S, Venance L. Endocannabinoids – LTP mediated by CB1 and TRPV1 receptors encodes for limited occurrences of coincident activity in neocortex. Front Cell Neurosci. 2018; 12: 182. doi 10.3389/fncel.2018.00182.
  51. Fatahi Z, Reisi Z, Rainer G, Haghparast A, Khani A. Cannabinoids induce apathetic and impulsive patterns of choice through CB1 receptors and TRPV1 channels. Neuropharmacology. 2018; 133: 75-84.
  52. Storozhuk MV, Moroz OF, Zholos AV. Multifunctional TRPV1 ion channels in physiology and pathology with focus on the brain, vasculature and some visceral systems. Biomed Res Int. 2019; Article ID 5806321. doi.org/10.1155/2019/5806321.
  53. Brito R, Sheth S, Mukherjea D, Ramkumar V. TRPV1: A potential drug target for treating various diseases. Cells. 2014; 3: 517-545.
  54. Yang F, Zheng J. Understanding spiciness: mechanism of TRPV1 channel activation by capsaicin. Protein Cell. 2017; 8: 169-177.
  55. Liu T-Y, Chu Y, Mei H-R, Chang D, Chuang H-H. Two vanilloid ligand bindings per channel are required to transduce capsaicin-activating stimuli. Fr Mol Neurosci. 2020; 12: 302. doi: 10.3389/fnmol.2019.00302.
 

Article Info

Article Notes

  • Published on: January 27, 2020

Keywords

  • Capsaicin

  • Pain
  • TRPV1
  • Rat Brain
  • CNS
  • Neurogenesis
  • Synaptic Plasticity
  • Histone Deacetylase (HDAC)2
  • Neuropsychiatric Disorders

*Correspondence:

Dr. Loris A Chahl
School of Biomedical Sciences and Pharmacy University of Newcastle NSW 2308 Australia
Email: loris.chahl@newcastle.edu.au.