GET THE APP

Hippocampal CA2 Region: A New Player in Social Dysfunctions

Journal of Neurology & Neurophysiology

ISSN - 2155-9562

Review Article - (2016) Volume 7, Issue 1

Hippocampal CA2 Region: A New Player in Social Dysfunctions

Xiang Mou*
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA
*Corresponding Author: Xiang Mou, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA, Tel: 713-798-5809, Fax: 713-798-5765 Email:

Abstract

For social species, normal social cognitive functions are essential for individuals to survive in a social group. Various neuropsychiatric disorders, including schizophrenia, autism and bipolar disorder are characterized by the impairments in social cognition. The hippocampus has been at the forefront of research in learning and memory for several decades. Hippocampal dysfunction contributes to learning and memory impairments as well as a range of social dysfunctions. However, precise contributions of different hippocampal sub-regions to differential aspects of cognitive functions remain unclear. This review first describes several key features of social abnormalities related to neuropsychiatric disorders, and then introduces the behavioral tasks used in animal models to assess each feature. Second, it gives a basic description of CA2 anatomy followed by an overview of its known functions. Third, it highlights a number of recent findings that associate the abnormalities of the hippocampal CA2 area and social dysfunctions in both animal models and human patients.

Keywords: Hippocampus, CA2, Memory, Schizophrenia, Autism, Social dysfunction

Introduction

Many animals, including humans, are social species. Social cognitive functions, such as social interaction and social memory, enable individuals to survive in a social group. Impairments in these functions are prominent traits in major neuropsychiatric disorders, such as schizophrenia, autism and bipolar disorder. Despite recent advances in the etiology of these disorders, the genetic and physiological underpinnings are still poorly understood.

The hippocampus is one of the most thoroughly studied structures in the brain, and has long been considered the hub of learning and memory. Hippocampal dysfunction contributes to learning and memory impairments observed in a range of neuropsychiatric disorders [1-6]. However, precise contributions of different hippocampal subregions to the maintenance of normal cognitive function remain unclear. Increasing evidence indicates that CA2, a small area interposed between CA3 and CA1 in hippocampus, plays an important role in pathology of some neuropsychiatric disorders.

In the present review, author first describes several major social abnormalities related to neuropsychiatric disorders, and how to assess those using rodent models. Second, author will provide the description of CA2 anatomy and its known functions. Finally, author will review some evidence that link CA2 to neuropsychiatric disorders.

Manifestation and assessment of social dysfunctions in animal models

Social dysfunction is a hallmark of various psychiatric diseases. Although considerable efforts are focused on understanding the genetic causes, and tremendous progress has been made, diagnosis of social dysfunction is still based on several categories of behavioral criteria. For example, the World Health Organization requires the presence of three key elements on diagnosing autism spectrum disorder (ASD): reduced social interactions, impaired communication and stereotyped behaviors [7]. Social dysfunctions can be characterized by, although not limited to, these three key features. Developing animal models to assess the social dysfunctions mentioned above has been challenging, since symptoms may be unique to humans and are often variable between human patients. Nonetheless, behavioral neuroscientists have made great efforts to develop standardized assays for social dysfunctions in animal models. Author will review assays that address aforementioned three aspects of social dysfunctions individually.

In humans, abnormal social interactions manifest in lack of interest in peers, failure to maintain eye contact and meaningful social interaction. In rodents, the assays of social interaction parameters include nose-to-nose sniffing, chasing, pushing, mounting in group social settings [8-12]. Such parameters can be scored manually or automated off-line by using video tracking system. More targeted measures of social interaction can be achieved in a three-chamber assay where an animal is given free choice to interact with a novel object or a novel conspecific, that are constrained in two separated chambers (social approach) [13-16]. The apparatus in this task is designed to allow transmission of social cues, such as visual, olfactory, auditory and tactile contact between two animals but no aggressive and sexual interactions. Wild type subjects spend more time in the chamber containing the novel conspecific than in the chamber containing the novel object, whereas subjects with social dysfunctions display otherwise. The three-chamber assay can also be used to test an animal’s social recognition: the ability to recognize familiar versus novel conspecifics. Rodents have natural inheritance to explore and interact with novel conspecifics versus familiar conspecifics, presumably due to their intact social memory. The social recognition or social memory is measured by the amount of time the subject spend with a novel conspecific versus a familiar conspecific [17-20]. Thus, in both assays wild type subjects tend to spend more time in the chamber with a novel conspecific.

Unlike humans, rodents do not rely heavily on language to communicate. Among the sensory cues rodents use to communicate, olfactory cues are of critical importance. Interestingly, interaction with cagemates who have just consumed novel flavored food resulted in the subject choosing food of the same flavor in preference to a completely novel flavored food [19,21-23], suggesting that rodents exchange meaningful information through olfactory signals. Pheromones are another important type of sensory cue that rodents relay upon. Mice and rats mark their territories with urinary scent as well as explore urinary scent marks in a cage and sniff urine soaked cotton swabs [24,25]. Olfactory communication can be quantified by scoring subjects’ interaction with pheromones compared to a non-biological scent. Pheromones elicit higher levels of sniffing than non-social odors [19,26].

Human repetitive behaviors include motor stereotypies, repetitive use of objects, resistance to change, and a narrow range of interests. Wide-type rodents exhibit spontaneous motor activities, including climbing, jumping and self-grooming. Repetitive behavior in rodents is usually measured by repetitive self-grooming. Mouse models with social dysfunction may self-groom for up to 2 minutes at a time, whereas wild type control mice generally groom for just several seconds [27]. Marble burying is another type of repetitive behavior [28]. Experimenters score the repetitive digging behavior by counting the remaining unburied marbles. Both behaviors can be scored manually or automated by software off-line. Other tests of repetitive behavior may include utilizing a standard T-maze. In this case, the animals are tested on their flexibility of learning. Animals are required to find food reward placed in the opposite arm of the T-maze from where they first learned to find the reward, which is referred to as reversal learning task [13,29,30]. An autism mouse model performed well on the initial acquisition stage but failed on reversal learning [31].

CA2 as a functional hippocampal sub region

Since the famous 1957 case study of patient H.M., who lost the ability to form anterograde memories after surgical removal of the hippocampus to treat epilepsy, the hippocampus has been at the core of research in bases of memory and memory-related neurological disorders. Extensive research led to the discoveries of neurophysiological mechanisms of memory at both cellular and circuit level, and detailed depiction of anatomical connectivity within hippocampus. The classic view of hippocampal anatomy divides hippocampus into three major subdivisions: dentate gyrus (DG), CA3 and CA1, each plays key roles in different aspects of learning and memory. Recently, the role of a long-neglected subdivision of the hippocampus, CA2, has come to light. An increasing body of evidences suggests that CA2 itself is far more than a passive transition region between CA1 and CA3 but a functional unit [32-40].

Anatomical and neurophysiological evidences have pointed to CA2 as the only subregion in the hippocampus where spatial learning components within hippocampus-surrounding areas (including entorhinal cortex, subiculum, parahipocampal areas) converge [33]. So how does CA2 integrate the differential incoming cues routed by distinct learning components? Several interventional studies have shed some light on this question. Knockout of vasopressin 1b receptor (V1bR) in mouse CA2 resulted in impairment in tasks that require remembering the temporal order of presented objects [41]. Recently, in vivo recording in behaving rats demonstrates that individual CA2 cells display standard firing patterns as in CA1 and CA3. However, CA2 differs from the other two subregions in that ensemble of CA2 cells do not maintain their spatial coding over time [36]. Taken together, these studies suggest that CA2 may participate in associating spatial and temporal information during learning, yet absent for memory consolidation.

Link between CA2 and social dysfunctions

Hippocampal dysfunction contributes to learning, memory and social impairments in a range of neuropsychiatric disorders, but the precise contributions of different hippocampal subregions remain largely unclear. As mentioned above, CA2 cells have weak coding for space, but what is their role in encoding emotional and social aspects of cognition? Recent discoveries extend our understanding of the association between CA2 and social functions, particularly social memory.

Gene expression profiling revealed that a number of social abnormality-related genes are preferentially expressed in CA2. Vasopressin 1b receptor (V1bR), one principal receptor mediating social aggression in rodents, was found prominent expression within the CA2 pyramidal cells but much lower expression in the hypothalamic paraventricular nucleus and amygdala [41]. Knockout of vasopressin 1b receptor in mouse CA2 reduces aggression and impairs social recognition, but spares spatial learning [42]. The adenosine A1 receptor has strongest immunoreactivity in adult rat CA2 [43]. mRNA expression of a neurotrophic factors, fibroblast growth factor (FGF-2), increases only in CA2 after controlled stress [44]. In addition, a G-protein signaling component, RGS14, is highly enriched in CA2 pyramidal neurons and plays a role in regulating synaptic plasticity [35].

Hitti and Siegelbaum showed that genetic inactivation of CA2 pyramidal cells in adult mice led to pronounced impairment of social memory [32]. Animals with CA2 inactivation lost the ability to recognize a familiar conspecific, thus spending same amount of time interacting with the familiar conspecific in a three-chamber social novelty test. This was not due to the loss of general memory function in these mice since they performed normally in several other hippocampus-dependent behaviors. Reversely, in a separate study, excitation of vasopressin 1b receptors (Avpr1b) in CA2 markedly enhanced social memory in mice during acquisition, but not during retrieval [38]. This enhancement can be blocked by Avpr1b antagonist. In Avpr1b knockout mice, lentiviral delivery of Avpr1b into the dorsal CA2 restored the social attack behavior [45]. Furthermore, a lesion study confirmed that specific excitotoxic NMDA inactivation of CA2 impaired social recognition in mice [39].

Several indications exist that CA2 pathology is a potential substrate for a number of neuropsychiatric disorders. Piskorowski et al. [37] reported that in the 22q11.2 mouse model (a mouse model for neuropsychiatric disorders, including autism and schizophrenia), the density of parvalbumin-expressing interneurons was decreased specifically in CA2 of adult mice. CA2 pyramidal neurons displayed a more hyperpolarized resting membrane potential, which made them less responsive to input stimulation. Social memory was also impaired in this mouse line assessed by direct interaction test. Similarly, reduction in parvalbumin immunoreactivity in CA2 [46,47], as well as AMPA [48] and histamine H3 receptors [49], was also observed in schizophrenia patients. Also in the 22q11.2 mouse line, Takahashi et al. demonstrated that in contrast to wild-type mice, heterozygous pups used invariable vocal sequences with less complicated call types, which resulted in inefficient maternal approach [50]. This suggests that neonatal maternal care and social communication play a critical role in the development of normal social functions in later adult life. Further studies may help pinpoint the neural substrate of the neonatal social communication inefficiency, which is absent in this study. In Parkinson’s disease patients, the elevated expression of amyloid beta peptide in CA2 was associated with dementia [51].

Conclusion

Social cognition is a complex behavior. Intact social memory and communication ability are essential for this process. Functional deficits in social memory underscore various neuropsychiatric disorders. Different hippocampal subregions collectively serve as fundamental substrates of cognitive functions, particularly learning and memory. Among them, the CA2 area displays distinct neurochemical and structural features than the others. Recently, it has been revealed that the CA2 mediates social recognition and plays an important role in social memory formation. Compromising CA2 by genetic and pharmacological manipulations results in disruptions of social cognitive functions. Recently, brain-derived neurotrophic factor (BDNF) has been suggested to play an important role in neuropsychiatric diseases, including Schizophrenia. BDNF mRNA and proteins are expressed in CA2 [52-54]. BDNF is essential for neuronal plasticity, a functional necessity for learning and memory. Therefore, deficits in BDNF signaling as well as BDNF mRNA polymorphisms may have indications in social dysfunctions observed in neuropsychiatric disorders [55,56].

While rodents and humans do not share the full spectrum of social dysfunction features, investigating social dysfunctions in animal models and identifying common neural mechanisms are essential from both scientific and clinical perspective. In that regard, elucidating the functions CA2 play in social cognition deficits will help pave the road for future treatment evaluation and drug development.

References

  1. Ahern TH, Modi ME, Burkett JP, Young LJ (2009) Evaluation of two automated metrics for analyzing partner preference tests. J Neurosci Methods 182: 180-188.
  2. Arakawa H, Arakawa K, Blanchard DC, Blanchard RJ (2008) A new test paradigm for social recognition evidenced by urinary scent marking behavior in C57BL/6J mice. Behav Brain Res 190: 97-104.
  3. Autry AE, Monteggia LM (2012) Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol Rev 64: 238-258.
  4. Bakker J, Honda S, Harada N, Balthazart J (2002) Sexual partner preference requires a functional aromatase (cyp19) gene in male mice. HormBehav 42: 158-171.
  5. Balu DT, Lucki I (2009) Adult hippocampal neurogenesis: regulation, functional implications, and contribution to disease pathology. NeurosciBiobehav Rev 33: 232-252.
  6. Blanchard DC, Blanchard RJ (1988) Ethoexperimental approaches to the biology of emotion. Annu Rev Psychol 39: 43-68.
  7. Bland ST, Tamlyn JP, Barrientos RM, Greenwood BN, Watkins LR, et al. (2007) Expression of fibroblast growth factor-2 and brain-derived neurotrophic factor mRNA in the medial prefrontal cortex and hippocampus after uncontrollable or controllable stress. Neuroscience 144: 1219-1228.
  8. Bolivar VJ, Walters SR, Phoenix JL (2007) Assessing autism-like behavior in mice: variations in social interactions among inbred strains. Behav Brain Res 176: 21-26.
  9. Chadman KK, Gong S, Scattoni ML, Boltuck SE, Gandhy SU, et al. (2008) Minimal aberrant behavioral phenotypes of neuroligin-3 R451C knockin mice. Autism Res 1: 147-158.
  10. Chen G, Chen KS, Knox J, Inglis J, Bernard A, et al. (2000) A learning deficit related to age and beta-amyloid plaques in a mouse model of Alzheimer's disease. Nature 408: 975-979.
  11. Conner JM, Lauterborn JC, Yan Q, Gall CM, Varon S (1997) Distribution of brain-derived neurotrophic factor (BDNF) protein and mRNA in the normal adult rat CNS: evidence for anterograde axonal transport. J Neurosci 17: 2295-2313.
  12. DeCarolis NA, Eisch AJ (2010) Hippocampal neurogenesis as a target for the treatment of mental illness: a critical evaluation. Neuropharmacology 58: 884-893.
  13. DeVito LM, Konigsberg R, Lykken C, Sauvage M, Young WS, et al. (2009) Vasopressin 1b receptor knock-out impairs memory for temporal order. J Neurosci 29: 2676-2683.
  14. Ferguson JN, Aldag JM, Insel TR, Young LJ (2001) Oxytocin in the medial amygdala is essential for social recognition in the mouse. J Neurosci 21: 8278-8285.
  15. Galef BG (2003) Social learning of food preferences in rodents: rapid appetitive learning. CurrProtocNeurosci Chapter 8: Unit 8.
  16. Gao XM, Sakai K, Roberts RC, Conley RR, Dean B, Tamminga CA (2000) Ionotropic glutamate receptors and expression of N-methyl-D-aspartate receptor subunits in subregions of human hippocampus: effects of schizophrenia. Am J Psychiatry 157: 1141-1149.
  17. Harrisberger F, Smieskova R, Schmidt A, Lenz C, Walter A, et al. (2015) BDNF Val66Met polymorphism and hippocampal volume in neuropsychiatric disorders: A systematic review and meta-analysis. NeurosciBiobehav Rev 55: 107-118.
  18. Hitti FL, Siegelbaum SA (2014) The hippocampal CA2 region is essential for social memory. Nature 508: 88-92.
  19. Hoeffer CA, Tang W, Wong H, Santillan A, Patterson RJ, et al. (2008) Removal of FKBP12 enhances mTOR-Raptor interactions, LTP, memory, and perseverative/repetitive behavior. Neuron 60: 832-845.
  20. Hsieh J, Eisch AJ (2010) Epigenetics, hippocampal neurogenesis, and neuropsychiatric disorders: unraveling the genome to understand the mind. Neurobiol Dis 39: 73-84.
  21. Jin CY, Anichtchik O, Panula P (2009) Altered histamine H3 receptor radioligand binding in post-mortem brain samples from subjects with psychiatric diseases. Br J Pharmacol 157: 118-129.
  22. Jones MW, McHugh TJ (2011) Updating hippocampal representations: CA2 joins the circuit. Trends Neurosci 34: 526-535.
  23. Kalaitzakis ME, Christian LM, Moran LB, Graeber MB, Pearce RK, et al. (2009) Dementia and visual hallucinations associated with limbic pathology in Parkinson's disease. Parkinsonism RelatDisord 15:196-204.
  24. Kaneko N, Sawamoto K (2009) Adult neurogenesis and its alteration under pathological conditions. Neurosci Res 63: 155-164.
  25. Knable MB, Barci BM, Webster MJ, Meador-Woodruff J, Torrey EF, et al. (2004) Molecular abnormalities of the hippocampus in severe psychiatric illness: postmortem findings from the Stanley Neuropathology Consortium. Mol Psychiatry 9: 609-620.
  26. Kohara K, Pignatelli M, Rivest AJ, Jung HY, Kitamura T, et al. (2014) Cell type-specific genetic and optogenetic tools reveal hippocampal CA2 circuits. Nat Neurosci 17: 269-279.
  27. Lagace DC, Donovan MH, DeCarolis NA, Farnbauch LA, Malhotra S, et al. (2010) Adult hippocampal neurogenesis is functionally important for stress-induced social avoidance. ProcNatlAcadSci U S A 107: 4436-4441.
  28. Lee SE, Simons SB, Heldt SA, Zhao M, Schroeder JP, et al. (2010) RGS14 is a natural suppressor of both synaptic plasticity in CA2 neurons and hippocampal-based learning and memory. ProcNatlAcadSci U S A 107: 16994-16998.
  29. Lim MM, Wang Z, Olazábal DE, Ren X, Terwilliger EF, et al. (2004) Enhanced partner preference in a promiscuous species by manipulating the expression of a single gene. Nature 429: 754-757.
  30. Mankin Emily A, Diehl Geoffrey W, Sparks Fraser T, Leutgeb S, Leutgeb Jill K (2015) Hippocampal CA2 Activity Patterns Change over Time to a Larger Extent than between Spatial Contexts. Neuron 85:190-201.
  31. McFarlane HG, Kusek GK, Yang M, Phoenix JL, Bolivar VJ, et al. (2008) Autism-like behavioral phenotypes in BTBR T+tf/J mice. Genes Brain Behav 7: 152-163.
  32. Moon J, Beaudin AE, Verosky S, Driscoll LL, Weiskopf M, et al. (2006) Attentional dysfunction, impulsivity, and resistance to change in a mouse model of fragile X syndrome. BehavNeurosci 120: 1367-1379.
  33. Moy SS, Nadler JJ, Perez A, Barbaro RP, Johns JM, et al. (2004) Sociability and preference for social novelty in five inbred strains: an approach to assess autistic-like behavior in mice. Genes Brain Behav 3: 287-302.
  34. Nadler JJ, Moy SS, Dold G, Trang D, Simmons N, et al. (2004) Automated apparatus for quantitation of social approach behaviors in mice. Genes Brain Behav 3: 303-314.
  35. Nagahara AH, Tuszynski MH (2011) Potential therapeutic uses of BDNF in neurological and psychiatric disorders. Nat Rev Drug Discov 10: 209-219.
  36. Ochiishi T, Saitoh Y, Yukawa A, Saji M, Ren Y, et al. (1999) High level of adenosine A1 receptor-like immunoreactivity in the CA2/CA3a region of the adult rat hippocampus. Neuroscience 93: 955-967.
  37. Pagani JH, Zhao M, Cui Z, Avram SK, Caruana DA, et al. (2015) Role of the vasopressin 1b receptor in rodent aggressive behavior and synaptic plasticity in hippocampal area CA2. Mol Psychiatry 20: 490-499.
  38. Rebecca PA, Nasrallah K, Diamantopoulou A, Mukai J, Hassan Sami I, et al. (2016) Age-Dependent Specific Changes in Area CA2 of the Hippocampus and Social Memory Deficit in a Mouse Model of the 22q11.2 Deletion Syndrome. Neuron 89: 163-176.
  39. Russo-Neustadt AA, Alejandre H, Garcia C, Ivy AS, Chen MJ (2004) Hippocampal brain-derived neurotrophic factor expression following treatment with reboxetine, citalopram, and physical exercise. Neuropsychopharmacology 29: 2189-2199.
  40. Smith AS, Williams Avram SK, Cymerblit-Sabba A, Song J, Young WS (2016) Targeted activation of the hippocampal CA2 area strongly enhances social memory. Mol Psychiatry.
  41. Stevenson EL, Caldwell HK (2014) Lesions to the CA2 region of the hippocampus impair social memory in mice. Eur J Neurosci 40: 3294-3301.
  42. Takahashi T, Okabe S, Broin PÓ, NishiA, Ye K,et al. (2015) Structure and function of neonatal social communication in a genetic mouse model of autism. Mol Psychiatry.
  43. Terranova ML, Laviola G (2005) Scoring of social interactions and play in mice during adolescence. CurrProtocToxicol Chapter 13: Unit13.
  44. Thomas A, Burant A, Bui N, Graham D, Yuva-Paylor LA, et al. (2009) Marble burying reflects a repetitive and perseverative behavior more than novelty-induced anxiety. Psychopharmacology (Berl) 204: 361-373.
  45. Wersinger SR, Ginns EI, O'Carroll AM, Lolait SJ, Young WS (2002) Vasopressin V1b receptor knockout reduces aggressive behavior in male mice. Mol Psychiatry 7: 975-984.
  46. Wesson DW, Keller M, Douhard Q, Baum MJ, Bakker J (2006) Enhanced urinary odor discrimination in female aromatase knockout (ArKO) mice. HormBehav 49: 580-586.
  47. Wetmore C, Olson L, Bean AJ (1994) Regulation of brain-derived neurotrophic factor (BDNF) expression and release from hippocampal neurons is mediated by non-NMDA type glutamate receptors. J Neurosci 14: 1688-1700.
  48. Wrenn CC (2004) Social transmission of food preference in mice. CurrProtocNeurosci Chapter 8: Unit 8.
  49. Wrenn CC, Harris AP, Saavedra MC, Crawley JN (2003) Social transmission of food preference in mice: methodology and application to galanin-overexpressing transgenic mice. BehavNeurosci 117: 21-31.
  50. Yang M, Clarke AM, Crawley JN (2009) Postnatal lesion evidence against a primary role for the corpus callosum in mouse sociability. Eur J Neurosci 29: 1663-1677.
  51. Yang M, Crawley JN (2009) Simple behavioral assessment of mouse olfaction. CurrProtocNeurosci Chapter 8: Unit 8.
  52. Yang M, Weber MD, Crawley JN (2008) Light phase testing of social behaviors: not a problem. Front Neurosci 2: 186-191.
  53. Yang M, Zhodzishsky V, Crawley JN (2007) Social deficits in BTBR T+tf/J mice are unchanged by cross-fostering with C57BL/6J mothers. Int J DevNeurosci 25: 515-521.
  54. Young WS, Li J, Wersinger SR, Palkovits M (2006) The vasopressin 1b receptor is prominent in the hippocampal area CA2 where it is unaffected by restraint stress or adrenalectomy. Neuroscience 143: 1031-1039.
  55. Zhang Z, Sun J, Reynolds GP (2002) A selective reduction in the relative density of parvalbumin-immunoreactive neurons in the hippocampus in schizophrenia patients. Chin Med J (Engl) 115: 819-823.
  56. (1992) World Health Organization. The ICD 10 Classification of Mental and Behavioural Disorder (Geneva, Switzerland).
Citation: Mou X (2016) Hippocampal CA2 Region: A New Player in Social Dysfunctions. J Neurol Neurophysiol 7:357.

Copyright: © 2016 Mou X. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.