Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Aug 1.
Published in final edited form as: Front Neuroendocrinol. 2010 Dec 13;32(3):303–319. doi: 10.1016/j.yfrne.2010.12.003

Influence of Photoperiod on Hormones, Behavior, and Immune Function

James C Walton 1,*, Zachary M Weil 1, Randy J Nelson 1
PMCID: PMC3139743  NIHMSID: NIHMS263912  PMID: 21156187

Abstract

Photoperiodism is the ability of plants and animals to measure environmental day length to ascertain time of year. Central to the evolution of photoperiodism in animals is the adaptive distribution of energetically challenging activities across the year to optimize reproductive fitness while balancing the energetic tradeoffs necessary for seasonally- appropriate survival strategies. The ability to accurately predict future events requires endogenous mechanisms to permit physiological anticipation of annual conditions. Day length provides a virtually noise free environmental signal to monitor and accurately predict time of the year. In mammals, melatonin provides the hormonal signal transducing day length. Duration of pineal melatonin is inversely related to day length and its secretion drives enduring changes in many physiological systems, including the HPA, HPG, and brain-gut axes, the autonomic nervous system, and the immune system. Thus, melatonin is the fulcrum mediating redistribution of energetic investment among physiological processes to maximize fitness and survival.

Keywords: Photoperiod, melatonin, immune, plasticity, seasonality, annual cycles, tradeoffs, life history strategy

1. Introduction

In order to be successful, individuals must grow, survive, and reproduce. Fitness is determined by how many offspring survive and ultimately produce offspring that themselves survive and reproduce. Investing in survival mechanisms depletes resources necessary for reproduction. Conversely, reproduction requires significant resources that may compromise survival. Thus, fitness reflects successful trade-offs between investments in the mechanisms underlying survival and reproduction that reflect life history strategies [291]. Natural selection has produced exquisite adaptations that have allowed individuals to successfully survive and reproduce in remarkably specific niches. Outside of the tropics, individuals have been selected to adapt to temporal niches, as well as spatial niches, because the yearly orbit of the Earth around the Sun drives seasonal variation in several environmental factors that affect temperature, weather, and food availability.

Habitats may vary substantially from winter to summer and in some cases distinctive sets of adaptations have evolved to cope with the often unique demands of winter and summer on survival and reproduction. For example, the winter set of adaptations may include a shift of energy allocations from non-essential functions such as growth and reproduction to those functions that are critical for immediate survival [82]. When the odds of successful reproduction are low, resources are shunted from reproduction and growth into survival mechanisms such as immune function, thermoregulation, or cellular maintenance. Consequently, over evolutionary time, seasonal patterns in the expression of adaptations have emerged that allow redistribution of energy resources to mediate trade-offs between traits such as immune function and reproductive effort [219]. During the fall and winter, the dual challenges of limited food availability with the need for additional energy to support thermogenesis make reproductive efforts unlikely to be successful, especially among small vertebrate animals; thus, these animals often reduce the size and function of their reproductive system [220]. In addition to reproduction, small nontropical vertebrates also display seasonal adjustments in body mass, adiposity, foraging, gut efficiency, pelage, sleep, growth, immune function, as well as cognitive and affective responses (reviewed in [253]). Because these seasonal adaptations often require significant time to develop, individuals rely upon an environmental signal to alter gene expression in order to produce the suite of season-specific adaptations.

Photoperiodism is the ability of plants and animals to measure environmental day length (photoperiod), a process that underlies the so-called biological calendar [225]. The biological ability to measure day length permits organisms to ascertain the time of year and engage in seasonally appropriate adaptations. Although the specific mechanisms that underlie the ability to measure day length differ among taxa, individuals that respond to day length can precisely, and reliably, ascertain the time of year with just two bits of data: (1) the length of the daily photoperiod, and (2) whether day lengths are increasing or decreasing. For individuals of many species, the annual cycle of changing photoperiod provides the environmental switch between seasonal phenotypes. Changes in day length, while probably of little direct importance to most animals, provide the most error-free indication of time of year, and thus enable individuals to anticipate seasonal conditions. Because the same photoperiod occurs twice a year (e.g., 21 March and 21 September), animals must be able to discriminate between these two dates; many photoperiodic vertebrates have solved this problem by developing an annual alteration between two physiological states (reviewed in [253]). Obviously, the environmental switch that controls the phenotypic trajectory is important in teasing out the interaction between environment and genes which drives phenotype.

The goal of this review is to focus on the influence of photoperiod on phenotype, the distribution of energetically expensive processes across the year to maximize survival and fitness, and the enduring effects of early life photoperiod on adult phenotype. Phenotype is the result of the interactions between genes and environment. In the wild, day length often determines the phenotype of newborn small vertebrates [46, 99, 129]. In mammals, photoperiodic information can even be passed to developing fetuses in utero so that the summer or winter phenotype can begin to develop prior to birth [323]. Thus, photoperiodic rodents born in the spring will grow to adult size, undergo puberty, and become reproductive in 6-8 weeks, whereas a sibling born in the autumn will not grow or undergo puberty for 4-5 months [108, 144]. Day length can be used in the laboratory as a precise environmental factor to probe gene expression during phenotypic development.

2. The pineal gland, melatonin, and photoperiod

To understand photoperiodism, it is important to understand how animals keep time physiologically. The pineal gland, and its hormone melatonin, mediates photoperiodic time measurement in mammals [136, 195]. Pinealectomy blocks responsiveness to photoperiod in every mammalian species studied [138]. Information about environmental light arrives to the brain via the lateral eyes in mammals [228]. A nonvisual neuronal pathway, the retinohypothalamic tract, carries light information from melanopsin expressing retinal ganglion cells directly to the suprachiasmatic nuclei (SCN) of the hypothalamus [55, 187, 284]. The SCN are the primary mammalian biological clocks [275]. From the SCN, the main pathway for photoperiod information to the pineal gland is relayed through the paraventricular nucleus of the hypothalamus, leaving the brain through the intermediolateral cells of the upper spinal cord, and then through the superior cervical ganglion. Postganglionic noradrenergic fibers project back into the brain and innervate the pineal gland, where neural information is transduced into a hormonal message [60, 100]. Melatonin, an indole amine hormone secreted rhythmically by the pineal [9], can induce photoperiodic responses when exogenously administered in a number of ways [20, 116, 284] (Figure 1). Particular aspects of the responses observed following constant release implants, daily injections, or daily infusions of melatonin led to some early controversy regarding the significance of various temporal characteristics of melatonin secretion. Melatonin is normally secreted in a circadian fashion, with an extended peak occurring at night, and basal secretion during the day [9]. The duration of this nocturnal ‘peak’ varies inversely with day length in several species, including humans. Results obtained in sheep and in Siberian hamsters strongly favor the overriding importance of the duration of the melatonin peak to transduce photoperiodic information. When pinealectomized hamsters or sheep were infused with melatonin on a daily basis, the types of responses elicited depended on the duration, but not on the phase, of the daily infusion [20, 38, 56]. Data obtained from similar studies in several other mammalian species support the conclusions that the duration of melatonin is the critical physiological parameter providing photoperiod information [13, 266, 272].

Figure 1.

Figure 1

Photic input is transduced by melanopsin-expressing retinal ganglion cells via the retinohypothalamic tract (RHT) to the suprachiasmatic nuclei (SCN). Output from the SCN is relayed through the paraventricular nucleus of the hypothalamus (PVN), to the intermediolateral cells of the upper spinal cord (IML), then to the superior cervical ganglion (SCG), and postganglionic innervation of the pineal gland regulates melatonin synthesis. Pineal melatonin carries photoperiodic information to distributed systems throughout the body, where it acts both directly and indirectly to regulate endocrine, neuronal, immunological, and behavioral processes.

With the use of 2-[125I]iodomelatonin, melatonin binding sites have been discovered throughout the periphery and nervous system in vertebrates [93]. Among mammals, high melatonin binding is commonly observed in the pars tuberalis (PT) of the pituitary, the suprachiasmatic nuclei (SCN) and dorsalmedial nucleus of the hypothalamus, preoptic area, and area postrema [154, 210, 318, 348]. Lesions or pharmacological blockade of 2-[125I]iodomelatonin-binding sites in the mediobasal hypothalamus blocks the inhibitory effects of endogenous and infused melatonin on gonadotropin secretion [15, 199, 242]. Low densities of 2-[125I]iodomelatonin binding have been reported elsewhere throughout the central nervous system (CNS) and periphery [93], and are reported to have a wide range of functions (reviewed in [94], including modulation of visual function in the retina [10, 91], immunomodulation [131], and cerebrovascular physiology (reviewed in [74]).

Two mammalian G protein-coupled melatonin receptors, MT1 and MT2, have been identified based on their affinity for 2-[125I]iodomelatonin and classified via molecular cloning techniques [92, 262]. Several other receptors in this family have been characterized (Mellc [97], GPR50 [263]), as well as other proteins that bind melatonin (MT3 [231], RZR/ROR [25]). MT1 is the dominantly expressed subtype of melatonin receptor, and targeted deletion of the MT1 gene in Mus disrupts prolactin release from the PT by altering clock gene expression, implicating it in neuroendocrine regulation of reproduction (reviewed in [318]). Further evidence for MT1 being the putative receptor for neuroendocrine photoperiodic responses comes from two photoperiodic species which do not express MT2 receptors: Siberian hamsters [324] and sheep [238]. Thus, all photoperiodic responses in these species are mediated via the MT1 receptor [238, 242]. MT1 activation primarily inhibits adenylate cyclase and enhances phosholipase C activation [114], but also has other effects through various signal transduction pathways, depending on tissue (reviewed in [196, 261, 318]).

At the intracellular level, melatonin, via activation of cell surface G protein-coupled receptors, interacts with circadian clock genes in target tissues to transmit the circadian light signal [138, 155, 212, 281]. Mammalian cellular circadian clocks are tightly regulated by a suite of clock genes that generate a self sustaining cycle via transcriptional feedback loops with approximately a 24 hr period. Cellular circadian rhythms are generated by a molecular pacemaker involving dimerized CLOCK-BMAL transcription factors that promote expression of period (PER) and cryptochrome (CRY) proteins, which in turn feedback to inhibit CLOCK and BMAL activity [164, 295]. Melatonin influences the rhythmic expression of several clock genes [155, 184], except in the SCN, where compensatory mechanisms may mask melatonin receptor signaling [281]. Onset of darkness and onset of light differentially modulate clock gene expression patterns in the PT: increasing levels of melatonin induce Cry expression, while decreasing levels induce Per1 expression [184, 205]; thus, the molecular circadian clock in extra-SCN tissues can entrain to the light phase of the external environment using a hormonal signal. These effects are indeed melatonin dependent, as pinealectomy or deletion of the MT1 receptor abolishes these rhythms in the PT [204, 317].

3. Photoperiod and energy balance

As noted above, individuals of several photoperiodic species undergo a suite of adaptive responses when exposed to short day lengths, putatively to allocate energy resources among energetically expensive processes to survive the harsh days of winter and to maximize fitness. Because many of these adaptations require significant time to develop, anticipating the appropriate adaptive response using photoperiodic information to alter physiology and reproductive state is critical to maximize survival (and ultimately, fitness) across seasons.

3.1. Somatic responses to photoperiod

The majority of research on somatic responses to photoperiod in mammals has been conducted in small rodents, which use different strategies in their responses to short day exposure. Although many temperate rodents regress their gonads in response to short days (see discussion below on reproductive responses to photoperiod [217]), they differ in their body mass response. Two alternate strategies are used to survive the energetic bottleneck of winter: 1) gain body mass via increased adiposity [22, 54, 170, 214], or 2) decrease body mass via decreased adiposity and regression of other tissues [41, 68, 70, 142, 226, 292, 319]. Increased adiposity equates to elevated endogenous energy stores for use during times of reduced energy intake, and decreased body mass would equate to reduced energy requirement to support total body mass; both are energy conserving strategies [69].

3.1.2. Neuroendocrine mechanisms

One of the hallmarks of photoperiodic responses to short days in rodents is the reduction in circulating gonadal steroids due to the involution of the gonads (see discussion of photoperiodic effects on the HPG axis below). Although gonadectomy can recapitulate the appropriate short day responses in body mass and adiposity in many rodents that use both adaptive strategies (reviewed in [225]), further reductions in mass occur when gonadectomized animals are exposed to short days [319]. Additionally, gonadectomized animals maintained in short days will become photorefractory and return to long day body mass in the absence of gonadal steroids [143]. Thus, gonadal steroids alone are not the only factor contributing to photoperiodic mass regulation.

In many species circulating leptin, which is synthesized in white adipose tissue, correlates strongly with body fat content, suggesting that it is the signal for monitoring adiposity. Leptin binds to receptors (Ob-Rb) in the arcuate nucleus (ARC), which in turn sends projections of orexigenic peptides (neuropeptide Y (NPY), agouti-related protein (AGRP), anorexigenic peptides (pro-opiomelanocortin (POMC), and cocaine- and amphetamine-related peptide (CART) to various nuclei in the hypothalamus to control feeding behaviors potentially based on a seasonal adiposity “set-point” (reviewed in [141, 211, 312]). Although there are seasonal changes in leptin sensitivity [171, 306], individuals are not in negative energy balance during photoperiod-mediated weight loss [1]. Precisely how the hypothalamic set point is established for satiety is unknown [21]. However, establishing the set point may involve converging input on the gut-brain axis of photoperiodic information (pineal melatonin), leptin signaling from the ARC, and ascending short-term satiety information from brainstem nuclei [140].

Within the mediobasal hypothalamus, triiodothyronine (T3) has been implicated in regulation of food intake [166]. One of the primary loci of photoperiodic signaling of pineal melatonin to the neuroendocrine axis is in the mediobasal hypothalamus (see above). Indeed, melatonin-dependent photoperiodic variation in deiodinase expression regulates T3 levels in the mediabasal hypothalamus [18, 264, 343]. Hypothalamic T3 levels are controlled by expression levels of deiodinase II (DIO2), which converts thyroxine (T4) into the active form of T3, and deiodinase III (DIO3), which leads to the conversion of T3 and T4 into inactive forms [136]. DIO3 is upregulated in short days, whereas DIO2 is upregulated in long days, resulting in elevated hypothalamic levels of active T3 during long days (see below). The majority of research on the contribution of thyroid hormones to photoperiodism has been in regulation of reproduction (see below); thus, the role of thyroid hormones in photoperiodic regulation of body mass has not been fully investigated.

In addition to hormonal and behavioral regulation of body mass, central control over energy homeostasis and lipid mobilization is regulated by autonomic outflow. The autonomic nervous system (ANS) innervates both white adipose tissue and brown adipose tissue [19, 48, 139, 177]. Both branches of the ANS are dynamically regulated by photoperiod [329], and the sympathetic branch of the ANS plays an important role in lipid regulation and lipid mobilization. Melatonin receptors (MT1) are expressed in many distributed forebrain nuclei involved in sympathetic outflow, and timed infusion of melatonin into these nuclei, mimicking short day levels for 5 weeks, can elicit short day body mass responses in Siberian hamsters [177].

3.1.4. Enteric mechanisms

Photoperiodic changes in gut morphology have been observed in diverse rodent species such as prairie voles (Microtus ochrogaster) [126], cavies (Microcavia australis) [271], alpine marmots (Marmota marmota) [148], and deer mice (Peromyscus maniculatus) [122]. Short photoperiod-mediated increases of intestinal mass and surface area help maximize energy and nutrient absorption during times of low food availability [122, 298]. In addition to photoperiod-mediated morphological changes to the gut, there is a growing body of evidence demonstrating coevolution of commensal gut microbiota and host (reviewed in [182]), and variations in gut microbiota composition can regulate host adiposity independent of changes in food intake [11-12, 181, 308]. Gut bacteria can sense, respond to, and participate in neurotransmission and neuroendocrine signaling [63, 147, 189-191, 233]; thus it is possible that gut microbiota may interact with the neuroendocrine system to contribute to the regulation of photoperiodic body mass changes.

In Siberian hamsters, photoperiod alters gut bacteria composition independent of changes in food intake [17]. Exposure to short days decreases relative abundance of Proteobacteria, which are most efficient at harvesting energy from high fat foods [17]. In a laboratory setting, exposure to long days increases high fat diet preference in Siberian hamsters [102], which raises the possibility that the gut microbiota may interact with photoperiod to drive seasonally specific feeding behaviors; however, synergy among behavioral, enteric, and neuroendocrine systems with photoperiod remains largely unexplored.

3.1.5 Photoperiod and central energy conservation

The CNS is composed of the most energetically demanding cells in the body [3]. Minimizing CNS metabolic demands during times of restricted energy availability (viz., short days), without impairing CNS function to the point of impacting long term reproductive fitness and survival, presumably provides adaptive advantages [150]. Seasonal brain plasticity has been reported for all vertebrate taxa [303], and photoperiodic brain plasticity has been studied in birds, primarily in two circuits; the song control system and the hippocampus. In support of this hypothesis of short-day energy conservation, upon exposure to long days, brain volume and soma size increases in several nuclei of the song system (reviewed in [150, 201]). In white-footed mice (Peromyscus leucopus), exposure to short days decreases hippocampal volume, with concurrent impairment in hippocampal function (see learning and memory below, [259, 337]). Short day reductions in brain mass have also been reported in meadow voles (Microtus pennsylvanicus) [67, 111], bank voles (Myodes glareolus) [342], and shrews (Sorex araneus L.) [72, 341]. However, short day increases in hippocampal volume occur in several species of wild food-caching birds [280]. Total brain volume also increases among long-day squirrels (Sciurus carolinensis) [173]. Energetic investment in increased brain volume in short days, in these cases, may impart an adaptive advantage by increasing spatial memory needed to locate food caches, which outweighs the increased energetic demands of supporting the increased neuronal volume. Alternatively, these volume increases could simply be a result of increased hippocampal usage during caching behavior (see [173, 280]). However, the direct contribution of varying seasonal demands in energy homeostasis to the evolution of these adaptations remains unspecified.

3.2. Photoperiod and immune function

Photoperiodic plasticity in the reproductive system is mirrored by alterations in various aspects of immunological physiology. Short day lengths generally enhance many types of immune responses in the laboratory, although others specific immune defenses are inhibited [220, 222]. The working theory in our laboratory has been that enhanced immunological defenses in short day lengths serve to oppose the stressful effects of winter conditions including reduced food availability and increased thermoregulatory demands [224] (Figure 2 A, B, D). This phenomenon of seasonal plasticity in the immune system is highly conserved and has been observed across vertebrate taxa. Most aspects of cell mediated immune responses including lymphocyte proliferation to mitogens, delayed-type hypersensitivity responses, and overall numbers of circulating immune cells are increased by exposure to short day lengths [32, 44, 83-84] (Figure 2 C). Additionally, both natural antibodies and antigen stimulated antibody responses are boosted by long term housing in short day lengths [83, 85] (Figure 2D). In contrast, most aspects of the innate arm of the immune system including cytokine responses to inflammogens are inhibited by winter-like day lengths [33, 247].

Figure 2.

Figure 2

The interaction of energetic stressors and photoperiod on immune function in deer mice (Peromyscus maniculatus). Short day exposure buffers stress responses to 2-deoxy-D-glucose induced metabolic stress (A) and to thermoregulatory stress (B). Short day exposure also enhances splenocyte proliferation in metabolically stressed mice in response to an immune challenge with the mitogen concanavalin A (C), and enhances basal serum IgG levels when mice are held in warm (20°C) ambient temperatures (D). Endogenous bolstering of immune function, coupled with blunted corticosterone responsiveness to stressors, represent short day adaptations that underlie energetic tradeoffs across the year mediated by photoperiod. The short day blunting of corticosterone responses serves to preserve immune function in the presence of stressors, and by applying and environmentally relevant stressor in the lab, such as cold temperature, immune parameters do not differ from long day mice (D). (Figures modified with permission from: [80](A,C); [83](B,D))

Seasonal plasticity in the immune system raises an important conceptual question. Why should immune responses be seasonally variable at all? Given the ubiquity of parasites and other pathogens why are immune defenses not maintained at maximal levels all year round? The answer may relate to the energetic cost of maintaining and activating immune function. Maximally investing in the immune system is not possible in the face of other energetically demanding processes [45, 79, 161]. During the spring and early summer small mammals invest heavily in the behavioral and physiological costs associated with breeding and most immune defenses are reduced [222]. However, during winter when successful breeding is typically impossible energetic investments are biased towards the immune system [225].

3.2.1 Mechanisms

Photoperiodic adjustments in immune defenses are mediated almost entirely by the pineal melatonin rhythm. Melatonin is generally anti-inflammatory and immunoenhancing in most laboratory animal species [128, 193]. However, melatonin largely recapitulates the short day effect among photoperiodic mammals rather than being universally immunoenhancing [34]. However, the way in which melatonin alters immune responses appears to be related both to direct actions of melatonin on immune cells, as well as via prolonged systemic adjustments that are associated with longer-term exposure to short day lengths. Pinealectomy prevents short-day induced changes in immune responses and exogenous melatonin can induce the short day phenotype in animals housed in long days [34, 84, 346]. Short day patterns of most immune responses require extended time in short photoperiods or long term exposure to short day patterns of melatonin. In contrast to the effects of long term melatonin treatment, short term (2 weeks) lengthening of the endogenous melatonin rhythm is not sufficient to alter either febrile or antibody responses in Siberian hamsters [34, 84]. There is one notable exception: melatonin added to lymphocyte cultures can directly modulate proliferative responses and do so in a photoperiod-dependent manner [251-252]. In sum, most of the immune changes associated with short days require extended periods to develop.

Although melatonin signaling is necessary for photoperiodic adjustments in immune responses, it is difficult to parse the differential direct effects of melatonin on immune cells from indirect modulatory actions on other neuroendocrine axes including the systems that control gonadal and adrenal steroids. However, the hypothesis that melatonin acts on immune function via downstream hormonal effects has not been born out, as gonadectomy and steroid replacement has minimal effect on photoperiodic plasticity in the immune system [86, 245]. Further, although androgens tend to be immunosuppressive estrogens often have the opposite effect [162]. Therefore, the short day induced gonadal regression would likely induce opposite effects in males and females if sex steroids were directly responsible for photoperiodic adjustments in immune responses. In general, both males and females respond similarly to changes in day length [35]. Gonadal steroids probably serve to modulate the larger melatonin-mediated effects of day length rather than directly driving photoperiodic changes [243].

Glucocorticoid hormones are also unlikely to be the primary drivers of photoperiodic changes, as circulating glucocorticoids are not systematically related to photoperiodic changes in immune responses across species [260, 328, 331]. Basal glucocorticoids are not the only relevant parameter associated with HPA axis function and some studies have reported photoperiodic adjustments in glucocorticoid receptors and feedback parameters [257, 267]. Additionally, acute stress-induced glucocorticoids are greatly enhanced in short day Siberian hamsters and elevated glucocorticoids appear to drive enhanced delayed type hypersensitivity responses in this species [32]. As sex steroids, glucocorticoids appear to act as modulators rather than driving forces behind photoperiodic adjustments in immune responses.

Immune function, as stated above, is energetically costly; thus, individuals, especially individuals of small vertebrate species, attend to their energetic status when mounting immune responses. That is, when energy is readily available more resources can be devoted to host defense, whereas when energy is more limited, resources will be more tightly controlled [77]. One hormone that is both immunomodulatory and responsive to changes in energy balance is leptin. Leptin, a peptide produced almost exclusively in adipose tissues, closely tracks body fat in mammals that undergo seasonal changes in body mass (see above [269, 273, 336]). For instance, Siberian hamsters, which lose body mass and adiposity in short day lengths, reduce circulating leptin, as well as leptin and leptin receptor gene expression in short days [146, 202, 268]. Siberian hamsters housed in short day lengths reduce antibody responses to the protein keyhole limpet hemocyanin [130, 244]. Exogenous leptin blocks the short day-mediated reduction in immune responses but only when food is available ad libitum [90]. When leptin-induced increases in food intake were prevented by food restriction, leptin no longer blocked the short day decrement in immune responses [90]. Similarly, reductions in body fat reduce humoral immune responses, but this effect can be blocked with the administration of exogenous leptin [82, 88]. Interestingly, leptin induced enhancement of antibody production requires intact innervation of the spleen suggesting that autonomic inputs are involved (see below, [76]) and leptin only alters immune responses in vivo, but not in vitro, suggesting that the effects are not mediated directly on the leukocytes [75]. Leptin likely provides important information about adiposity and thus can modulate photoperiodic adjustments in immune responses.

The autonomic nervous system is also a potential intermediate mechanism for photoperiodic modulation of the immune system. The sympathetic nervous system contributes directly (and presumably indirectly via changes in adiposity) to photoperiod changes in immune responses. For instance, surgical extirpation of the adrenal medulla, the principal source of blood-borne catecholamines, inhibits antibody production, but only in long days. On the other hand, denervation of the sympathetic inputs to the spleen significantly attenuates antibody production, but only in short days [81]. Splenic norepinephrine content is increased by short day lengths in Siberian hamsters and this phenomenon appears to be at least partially responsible for the short-day induced reduction in lymphocyte proliferation in this species. Adding norepinephrine to lymphocyte cultures attenuates proliferation responses, but only in short day lengths [78]. Although not yet functionally studied, photoperiod also increases activity of the parasympathetic branch of the autonomic nervous system, which may also have immunomodulatory actions [329].

3.3. Photoperiod and reproduction

Maintenance and support of reproductive function in mammals is dependent upon activation of the hypothalamic-pituitary-gonadal (HPG) axis. Pulsatile gonadotropin releasing hormone (GnRH) secretion from the hypothalamus into the hypophyseal portal system stimulates releases of gonadotropins (luteinizing hormone – LH, follicle-stimulating hormone – FSH) from the anterior pituitary, which in turn supports development and maintenance of mature gonads [42, 179]. The HPG axis can be modified at multiple levels to regulate reproductive function, some of which are influenced by photoperiod and the influence of photoperiod can vary depending on previous photoperiod exposure.

3.3.1 Short-day breeders

Sheep (Ovis aries), in common with other ungulates and most large seasonally-breeding mammals, are so-called ‘short-day breeders’. Ewes have relatively long periods of gestation; mating typically occurs in the autumn and lambs are born and nursed in the spring when food and other conditions are most conducive for survival. As day lengths decrease in late summer, the rate of GnRH secretion increases, which eventually stimulates increased gonadotropin secretion that initiates reproductive function [340]. In Suffolk ewes, seasonal reproductive transitions appear to reflect changes in the responsiveness of the GnRH neurosecretory system to the negative feedback of estradiol. In common with other mammals, GnRH neurons in sheep do not express estrogen receptor alpha (ER) [340]. Although ∼50% of GnRH neurons in sheep express estrogen receptor beta (ER) [287], the role of these receptors in estrogen feedback regulation of GnRH signaling remains unidentified. However, ER may be the putative estrogen receptor for feedback regulation of GnRH secretion as targeted disruption of ER in female mice does not impair reproduction [169, 186]. The ultrastructure and synaptic inputs of GnRH neurons in the preoptic area of ewes during the breeding season receive more than twice the mean number of synaptic inputs per unit of plasma membrane than GnRH neurons in anestrous animals [340], thus the influence of estradiol on GnRH neurosecretory activity may be conveyed via converging afferents on GnRH neurons from: A) estradiol sensitive glutamatergic cells in the hypothalamus, BNST, and brainstem [241], B) BNST neurons receiving input from estradiol sensitive noradrenergic cells in the brainstem [239], and C) dopaminergic input from the A15 nucleus [58]. These seasonal alterations in synaptic input are independent of seasonal fluctuations of steroid concentrations as both intact and ovariectomized ewes bearing estradiol implants show changes in synaptic inputs onto GnRH neurons [340], but how photoperiodic information is relayed through these pathways remains unspecified. After mating season, sheep become photorefractory; i.e., short days lose their stimulatory effects and mating behavior wanes. Exposure to the long day lengths of summer is not necessary to re-establish responsiveness to short days, suggesting the presence of an underlying circannual cycle of photosensitivity [265].

3.3.2 Long-day breeders

Syrian, or golden, hamsters (Mesocricetus auratus), and Siberian hamsters (Phodopus sungorus), represent the most common mammalian models used in laboratory investigations of photoperiodism. Hamsters, in common with most small mammals, are so-called ‘long-day breeders’. Gestation is relatively brief in these animals; mating, pregnancy, and lactation occur during the long days of late spring and early summer. The minimum day length that supports reproduction is called the ‘critical day length’. Critical day length is not a fixed variable, but differs among populations of animals living at different altitudes and latitudes [49]. Adult male Syrian hamsters undergo gonadal regression when day lengths fall below 12.5 hours of light [98]. However, Siberian hamsters, which live at higher latitudes than Syrian hamsters, display a critical day length for reproductive function of 16 hours of light/day [96]. In addition to latitudinal variation in critical day length, manipulation of photoperiod history in the lab can also influence critical day length. Siberian hamsters held in long or short day lengths will show correct photoperiodic reproductive responses to opposite photoperiods if placed in intermediate photoperiods that differ by >1.5 hr from the initial photoperiod [246, 250]. It should be noted here that most laboratory studies of photoperiodism involve abrupt transfer to different day lengths, which does not reflect naturalistic conditions. Using simulated natural photoperiod (SNP) exposure, to mimic gradual incremental changes in day length, in Siberian hamsters has demonstrated that photoperiod history is critical for determining critical day length. Critical day length can be shifted up to 6 hours by manipulating the simulated natural photoperiod history [121]. Thus, early observations that latitude determines critical day length, while correct, should be interpreted as photoperiod history, which changes with latitude, is responsible for determining critical day length.

When Syrian hamsters are maintained in day lengths < 12.5 hours of light, blood concentrations of gonadotropins and sex steroid hormones decrease, accessory organ mass diminishes to about 10% of the original size, and reproductive behaviors stop (reviewed in [253]). Male hamsters remain reproductively quiescent for approximately 16-20 weeks, a period of time that roughly corresponds to the duration of short days experienced in the wild during autumn and winter. Hamsters are photorefractory during the recrudescent phase; i.e., gonadal condition becomes unlinked from photoperiodic inhibition. Photorefractoriness permits attainment of fully functional gonads in the spring before environmental photoperiods attain 12.5 hours, the day length necessary for gonadal maintenance in the autumn. This is an important adaptation that allows burrowing animals (that live in constant dark conditions) to anticipate spring conditions with the development of fully functional reproductive systems without long-day exposure. Once animals become photorefractory exposure to increasing day lengths initiates reestablishment of photoresponsiveness, but the process takes ∼10 – 15 weeks to complete after initiation [51, 159].

One of the mechanisms by which the nightly duration of melatonin secretion affects the reproductive system is by altering the steroid negative-feedback mechanisms in the hypothalamus and pituitary gland (reviewed in [117]). In long-day and short-day breeders, reduced secretion of pituitary LH and FSH during the non-breeding season results from increased sensitivity of the hypothalamic-pituitary axis to the negative feedback effects of gonadal steroid hormones [176, 296, 307]. Low concentrations of blood androgens are more effective at inhibiting post-castration elevations in pituitary gonadotropin secretion in hamsters and rams, when males are housed in short or long days, respectively. A return to the lower level of sensitivity returns the animals to a state of reproductive activity via increased pituitary hormone secretion. A similar phenomenon has been implicated in puberty, during which a prepubertal decrease in sensitivity to steroid negative feedback leads to increased secretion of LH and FSH and activation of the reproductive system [286].

In addition to changes in the sensitivity of the gonadotropin secretion system to gonadal steroid hormones, several steroid-independent mechanisms have also been implicated in the regulation of seasonal changes in the rate of gonadotropin secretion. In long days, female hamsters display ∼8-10-fold increase in baseline serum LH concentrations following ovariectomy, and LH concentrations can be returned to baseline by administration of estrogens. After several weeks of short-day exposure, female hamsters become anovulatory and serum LH concentrations are very low during most of the 24 h cycle; however, the anovulatory females display daily LH surges during the afternoon. This pattern of LH secretion is steroid-independent, as it continues following ovariectomy or adrenalectomy [40]. A steroid-independent, but melatonin-dependent, photoperiodic LH secretion pattern difference is also found in ewes. In ovariectomized ewes, LH pulse frequency is suppressed in long days and recovers to normal breeding season rate in short days. This difference is abolished by pinealectomy, but recovered with melatonin infusions mimicking short- and long-day photoperiods [39]. Although it is currently unknown whether steroid-dependent or -independent mechanisms are more common in photoperiodic mammals, seasonal variation in circulating and pituitary concentrations of gonadotropins has been reported for several mammalian species [36, 39, 158, 203].

Recent research in birds has identified two members of the RFamide family of peptides that regulate the HPG axis, kisspeptin and gonadotropin inhibitory hormone (GnIH), which may have similar roles in mammals (reviewed in [124, 304]). These peptides act in an antagonistic fashion on GnRH neurons; kisspeptin stimulates GnRH release [288] and GnIH inhibits GnRH release [305]. Although the role of these peptides in photoperiodic control of the HPG axis has not been completely specified, both cell types are responsive to melatonin. In cultured mammalian hypothalamic neurons, expression of kisspeptin is decreased and RFamide-related peptide (RFRP, the mammalian homologue of GnIH) expression is increased by melatonin [113]. Exogenous and endogenous melatonin drives GnIH release in birds [62, 310], and short photoperiod increases RFRP expression in hamsters [197, 237]. Consistent with the effects of melatonin on kisspeptin in rats [113], kisspeptin immunoreactivity in the anteroventral periventricular nucleus of the thalamus of Siberian and Syrian hamsters is reduced in short days [123, 283]. However, in the arcuate nucleus, kisspeptin immunoreactivity is elevated in short-day Siberian hamsters [125, 198], but no effect of photoperiod was reported on gene expression [237].

As noted above, thyroid hormones have also been implicated in photoperiodic regulation of the HPG axis in both mammals and birds [107, 216, 347]. Hypothalamic T3 levels are higher in long days and hypothalamic implants of T3 can block short day photoperiodic responses in Siberian hamsters [18]. Exposure to long days increases thyroid stimulating hormone (TSH) release from the PT, and the increased TSH release from the PT acts in a paracrine manner to induce DIO2 expression in the tanycytes lining the third ventricle in the mediobasal hypothalamus, resulting in elevated T3 availability [134, 345]. Reciprocal expression of DIO2 and DIO3 in the mediobasal hypothalamus thereby regulates thyroid hormone activity in the hypothalamus, and expression of these genes is regulated by melatonin (see discussion above [232, 264, 322, 344-345]). Conservation of this mechanism suggests that it may represent an ancestral mechanism of seasonal timing driven by photoperiod [136-137].

4. Photoperiod, affect, and non-reproductive behaviors

Although many seasonal behaviors in photoperiodic mammals are specifically associated with reproduction (see above), there are several behavioral traits, such as affective responses and aggression, that are modulated by photoperiod independent of gonadal steroids. Immunological, hormonal, and neural factors, all of which are modulated by photoperiod, converge to modify behavioral output. Affective disorders are generally considered maladaptive; however, photoperiodic changes in affect may represent an adaptive strategy to conserve energy during the energetic bottlenecks encountered during the short days of winter (e.g., [229-230, 326]). Additionally, conservation of energy during the short days of winter may also be the putative mechanism driving photoperiodic modulation of aggression (see below). Not only are these traits influenced by photoperiod in mammals that respond reproductively to photoperiod, behavioral responses to photoperiod are also observed in mammals that do not respond reproductively to photoperiod, which has implications for developing animal models of human pathologies with a seasonal component, such as seasonal affective disorders and depression [338].

4.1. Affective responses

Observable and abnormal emotional states, such as excessive elation or sadness, define affective disorders [270]. Behavioral responses which comprise human affective disorders, such as altered food intake and reduced motivation (depressive-like), and increased anxiety and fearfulness (anxiety-like), can also be observed in rodents in a laboratory setting. Until recently, there was little evidence that photoperiod itself could alter affective behaviors, even though it has been known for decades that there is a correlation between season and affective disorders in humans [106, 180, 234]. Recent research has now identified several rodent species that display changes in affective behaviors that are induced by changes in photoperiod.

In reproductively photoperiodic rodents, exposure to short days induces changes in affective behaviors that are independent of changes in reproductive hormones. Exposure to short days increases anxiety-like and depressive-like responses in collared lemmings [327] and Siberian hamsters [249, 258]. These photoperiodic behavioral changes develop early during short day exposure [249], and can persist after maximal gonadal regression [258, 327], supporting the idea that the influence of photoperiod on affect can be independent of circulating gonadal steroids. Further support of this idea comes from a growing body of evidence showing that animals that do not respond reproductively to photoperiod also display photoperiodic responses [218, 293]. Short-day exacerbation of depressive-like and anxiety-like behaviors have been reported in both nocturnal rodents (rats [29, 207, 248]), and diurnal rodents (e.g., Nile grass rats [6], and sand rats [7-8]). These affective responses to short days are unambiguously linked directly to pineal melatonin secretion duration [8], which may model the extended duration of melatonin secretion observed in humans with seasonal affective disorder [326].

4.2 Non-reproductive social behaviors

It is presumably adaptive for organisms to differentially display complex social behaviors such as aggression and non-reproductive affiliation at different times of the year. Aggressive behaviors occur because of competition for limited resources, whereas affiliative behaviors occur when long term survival probability is increased by sharing resources with conspecifics to liberate physiological resources for other processes. Both aggression and affiliation display seasonal variation, influenced by photoperiod, in reproductive and non-reproductive contexts.

Rodents, such as wood rats (Neotoma fuscipes) [52], rat-like hamsters (Cricetus triton) [349], Siberian hamsters [87, 151, 276], California mice (Peromyscus californicus) [282], and Syrian hamsters [14, 103, 112, 152], increase aggression in short days. Elevated aggression in short days may be adaptive to protect limited resources or territories during the winter non-breeding season [87, 274, 297]. In males, testosterone concentrations are positively correlated with aggression during the breeding season [172, 221, 334]. Elevated aggression in short days, however, is typically independent of testosterone [52, 87, 112, 151-152, 349]. Although male aggression has been studied extensively, female aggression has received far less scientific scrutiny [227]. Nonetheless, elevated non-reproductive aggression in females also varies in response to photoperiod in rat-like hamsters [349], Siberian hamsters [276], California mice [282], and Syrian hamsters [103].

The precise mechanisms underlying short-day increases in aggression in both sexes remain unidentified; however, recent studies have identified some potential factors. Nitric oxide, a gaseous neurotransmitter, has been implicated in male aggression [223]. Short photoperiod reduces nNOS expression, the enzyme producing nitric oxide in neurons, in the amygdala of male Siberian hamsters, and nNOS expression is negatively correlated with aggression in short days [332]. Melatonin, a physiological signal of photoperiod, may directly modulate aggression. Laboratory house mice (Mus musculus) display increased territorial aggression when given a short day like regimen of melatonin [236]. Short-day alterations in aggression have been directly linked to pineal melatonin in Syrian hamsters [14, 103], Siberian hamsters [277], and fat sand rats [8]. Although testosterone concentrations are at their nadir in short days (see above), it is possible that testosterone synthesized and converted to estrogen de novo in the brain can influence short-day aggression [289]. Photoperiod and estrogen receptor distribution also interact to modulate short-day aggressive behaviors [301-302], but how activation of this pathway occurs in short days remains unspecified. Melatonin can also interact with the HPA axis, which has been implicated in the modulation of aggression [132]. Exposure to short days alters HPA axis reactivity [257] and the HPA axis interacts with melatonin to modulate aggression [87, 236].

In contrast to increased aggression, some rodent species display reduced aggression in short days [4, 8, 26]. To reduce thermoregulatory demands during the harsh days of winter, several rodent species form communal nests [4-5, 192, 335]. Conspecific tolerance in these communal nests necessarily requires reduced territorial aggression and increased affiliative behavior. The oxytocin (OT) and vasopressin (AVP) family of neuropeptides has been implicated in the control of diverse social behaviors, including affiliation and aggression (reviewed in [119, 315]). Generally in mammals, increased oxytocin binding is associated with increased affiliative behavior [175] and increased vasopressin binding is associated with aggressive behavior [31, 101]; thus, this system is poised to be a target for photoperiodic modulation of these behaviors. Indeed, pineal melatonin can modulate neurohypophysial AVP and OT neuronal activity (reviewed in [156]). Photoperiodic differences in oxytocin receptor distribution underlie increases in affiliation in short-day female meadow voles [28, 235], but the role of vasopressin in mediating photoperiodic changes in aggression are less clear (see [2, 37, 53]). Thus, although photoperiod and pineal melatonin modulate non-reproductive behaviors, the systems are distributed and the effects are species-specific.

4.3 Learning and memory

In addition to the social and affective behavioral changes described above, photoperiod can modulate learning and memory, potentially a functional result of altered hippocampal morphology (see above). In rodents, seasonal variation in spatial learning and memory has been reported in fox squirrels (Sciurus niger) [320], deer mice (P. maniculatus) [109-110], and white-footed mice [257, 259, 337]. In a naturalistic setting, seasonal variation in hippocampal and brain volume is positively correlated with seasonal spatial navigation and spatial memory requirements [173, 342]. Indeed, in deer mice, seasonal territory maintenance life history positively correlates with photoperiodic variation in spatial learning and memory, leading to sex and population based differences in spatial learning and memory performance across seasons [109-110]. When male white footed mice, which have seasonal variation in territory size, are brought into the lab where there are no territories to maintain, a similar decrease of hippocampal volume observed during winter in the wild is observed in laboratory short days [259], indicating an underlying photoperiodic mechanism in brain morphological plasticity. Supporting this conjecture, exposure to short days alone impairs spatial learning and memory, alters dendritic morphology of hippocampal neurons [259], and impairs long term potentiation [321], the putative mechanism for memory formation in the brain [43].

5. Enduring effects of photoperiod

5.1. Early life photoperiods organize adult phenotype

In addition to the seasonally variable phenotypic changes discussed above, photoperiod history can also affect adult phenotype. Many adult diseases and disorders are influenced by the season of birth. For example, season of birth in humans has been correlated with a number of disorders and pathologies, including increased risk of several types of cancer [23, 194], susceptibility to pulmonary fibrosis [133], metabolic disorders [127, 174, 311], and susceptibility to cardiovascular disease [47, 174]. Late gestation and birth in short days, in both northern and southern hemispheres, is associated with increased prevalence of schizoaffective disorder, autism, and major depression in the adult population [135, 299]. Season of birth may also differentially affect subtypes of affective disorders, as long day births are associated with an increase in the deficit subtype of schizophrenia [160] and risk of completed suicide [89]. Some of these disorders can be modeled by early life differences in photoperiod.

5.1.1. Somatic and reproductive effects of early life photoperiod

As described, the endogenous developmental programming effects of birth season can be investigated by manipulating an environmental factor (day length) that individuals of many animal species use to predict challenging conditions. Although much research on seasonality in small mammals has focused on photoperiod manipulations in adults, early-life photoperiod is also an important source of seasonal information and can establish individuals' developmental trajectory by regulating somatic and reproductive development [50]. Because the adaptations associated with investing in reproduction or survival mechanisms are generally mutually exclusive, it is useful for small animals to follow the appropriate seasonal developmental trajectory soon after birth. Siberian hamsters born late in the breeding season delay puberty until the following spring, whereas hamsters born early in the breeding season undergo rapid reproductive development, presumably to have the opportunity to mate prior to autumn [120]. Photoperiod information is communicated in utero to Siberian hamster pups [145] via the maternal melatonin rhythm. In addition to regulation of developmental processes, early life photoperiod also affects reproductive [145] and affective responses to day lengths later in life (discussed below). Intermediate day lengths (e.g., 14 h/day in Siberia) occur at both the beginning and the end of the breeding season; thus, the same day lengths forecast very different environmental conditions. Photoperiodic rodents solve this problem by comparing ambient day length to a reference day length that is encoded prenatally via a mechanism termed photoperiodic history [145, 246, 250, 279]. This mechanism appears to be mediated by melatonin rhythmicity. Pups born to pinealectomized dams are reproductively suppressed even when housed in long days presumably because the duration of the daily melatonin rhythm is increasing [309]. That is, pinealectomy of the dams blocks reception of any rhythmic melatonin signals in hamster fetuses and neonates. When the pups' circadian system matures such that the neuroendocrine system starts producing melatonin, despite encoding long days, this melatonin signal is interpreted as a decreasing photoperiod resulting in gonadal inhibition [309].

Photoperiodic rodents represent an ideal animal model for studying the programming, or organizational, effects of early life environmental conditions on adult phenotype. The effects of being born at different times of the year can be simulated in the lab by manipulating the perinatal and then postweaning photoperiod. As noted above, young animals are exceptionally responsive to day length information and early life photoperiod is sufficient to organize many physiological systems and establish a developmental trajectory. Male and female hamsters born in short days will delay reproductive maturation, but will rapidly undergo reproductive development if exposed to long days at weaning; the inverse case is also true [330]. In female hamsters, rearing in short days alters ovarian physiology through adult life, resulting in increased fecundity and fertility in old age, presumably due to the preservation of more primordial follicles [157]. Male Syrian hamsters that experience gestational short day exposure reduce peak testicular size compared to males gestated in long days [27].

Enduring effects of early life photoperiod are not limited to the reproductive system, however. Male meadow voles reared in short days have reduced brain mass as adults compared to long day reared counterparts [67]. Early removal of the pineal gland impairs antibody-dependent cellular cytotoxicity and alters thymic development in rodents [66, 316] and melatonin supplementation in ovo enhances cellular and humoral immune responses in domestic fowl [208]

5.1.2. Behavioral effects of early life photoperiod

Perinatal photoperiod conditions can organize adult affective behaviors and can interact with postweaning photoperiod conditions to regulate the expression of affective behaviors in adults. Hamsters exposed to short days early in life have increased anxiety- and depressive-like responses as adults [258]. Exposure to short days increase anxiety- and depressive-like behaviors in hamsters born in long days [249] and exposure to short days perinatally potentiates those differences in adulthood [258]. Additionally, postnatal pinealectomy, interpreted by the animal as a shift from short days to long days via decrease in melatonin duration (see above, [309]), leads to altered anxiety-like behaviors in adult Siberian hamsters [339].

As noted, enduring behavioral effects of early life photoperiod experience are not limited to rodents. Season of birth effects in humans may reflect endocrine changes in HPA axis responsivity. Within a patient population suffering from unipolar major depressive disorder, failure to suppress Cortisol production in the dexamethasone suppression test was significantly increased in patients born in winter [105]. In elderly outpatients in the southern hemisphere, being born during flu season in both hemispheres was correlated with increased depressive and suicidal symptoms [240]. Thus, factors other than the endocrine system (the immune system) may contribute to the enduring effects of photoperiod.

5.1.3. Immune effects of early life photoperiod

Immunologically, animals born at different times of the year permanently alter their immunological priorities to best meet the demands of the environment they are likely to experience. Male and female Siberian hamsters, maintained prenatally and until weaning (21 days) in either short days (8 h light/day; 8L:16D[SD]) or long days (16 h light/day; 16L:8D [LD]), were weaned into either the opposite photoperiod or maintained in their natal photoperiod, forming four groups (LD-LD, LD-SD, SD-LD and SD-SD). After 8 weeks in their respective postweaning conditions, cell-mediated immune activity was compared among groups. Enhanced delayed-type hypersensitivity (DTH) responses were observed in hamsters housed in short days both perinatally and postweaning relative to all other groups. Additionally, splenic masses were enhanced by perinatal and not postweaning exposure to short days. Thus, there are long-lasting organizational effects of perinatal photoperiod on the rodent immune system, but does this model the human state?

5.1.3.1. Season of birth effects on the immune system in humans

Across the globe, resource availability and environmental conditions vary in a predictable fashion over the course of a year. Pathogens, food availability, and ambient temperatures all vary seasonally. Therefore, organisms developing at different times of the year are likely to experience dissimilar environmental conditions. Studies examining season-of-birth effects on immune function have been relatively few, but best documented in humans. By examining epidemiological data for seasonal changes in immune-related disease states, it is possible to make inferences about the immune system. For example, in industrialized countries, the environment during early life has a profound effect on health outcome. Season of birth effects have been reported for cytokine responses [115] and numerous health conditions including asthma [118, 163], allergy [178, 314], inflammatory bowel disease [16, 206, 313], and multiple sclerosis [24, 200, 290, 300]. Although the directionality and strength of these relationships varies among the specific immunological component and disease state investigated, these season of birth differences are still apparent.

These seasonal patterns exist despite that most modern societies are buffered from much of the environmental variation that was presumably experienced by our ancestors [104]. In developing countries, the effects of early life experiences and environmental conditions are readily apparent. For example, in rural villages in the West African country of Gambia profound seasonality in humans remains extant. Despite being only 13° north of the equator, there are seasonal rhythms in rainfall that control the timing of the harvest. By the time the rains return in July, the previous year's stores of food have been exhausted. Therefore, there is a distinct season of restricted caloric intake that roughly coincides with the annual onset of the rains, termed the ‘hungry season’ [254]. Pregnant women reduce weight gain during that period [254]; in addition, the incidence of maternal and infant disease, mostly malaria and diarrhea, peak during this time [57]. Because there is a pronounced annual rhythm to these conditions, season of birth can be used as a proxy for early life experience of these factors. This seasonal phenomenon may have effects on immunological development. Children born during the hungry season have attenuated thymic development and are more likely to contract infections later as juveniles [64]. Perhaps the strongest evidence for a role of seasonality in the regulation of developmental processes and adult phenotype is that individuals born during the hungry season are significantly more likely to die prematurely than are those born when food is more plentiful [209]. Moreover, a disproportionate number of these early deaths are associated with infections [209]. It remains unclear whether the effects on phenotype in this population are due to direct actions of malnutrition and early life infection, or indirect endogenous changes in developmental trajectory in response to these conditions.

6. Epigenetic control of gene expression and photoperiodism

Mechanistically, it is unknown how early life photoperiod imparts its enduring effects on adult phenotype. It is possible that photoperiod-based epigenetic modifications occur in the germline cells, in utero, or in early life, that endure in the absence of the original causative signal. Although the broad use of the term “epigenetics” in recent literature to describe changes in gene regulation [256] strays from its classical definition as “a stably heritable phenotype resulting from changes in a chromosome without alternations in the DNA sequence” [30], we will use this term in the context of lasting changes in gene expression in the absence of the original causative signal [95]. Epigenetic chromatin modifications have become a focus of research as a putative mechanism for the long-term regulation of gene expression to evoke phenotype. Although detailed description of the mechanisms and molecular pathways involved in epigenetic regulation of gene transcription is beyond the scope of this review (for reviews see [61, 153, 255]), modifications of histone proteins lead to chromatin remodeling which either permits or prohibits RNA polymerase access to the DNA for transcription, leading to altered gene expression (phenotype) in the absence of genotypic change. These epigenetic modifications can occur as a result of response to an environmental factor, a behavioral factor, or via transmission through the germline.

6.1. Genomic imprinting

Genomic imprinting results from germline epigenetic modifications leading to monoallelic expression of genes from either the maternal or paternal line [95, 333]. Whereas over 100 imprinted genes have been identified in rodents and humans [95], the presence of over 600 autosomal imprinted genes, comprising 2.5% of the genome, was predicted for Mus musculus [188]. Among imprinted genes already identified are a number of genes directly impacting endocrine function, as well as neuropeptides in behavioral circuits such as the monaminergic system, GABAergic system, and the AVP/OT system (reviewed in [71]). Additionally, recent studies have identified changes in methylation patterns in immune pathways which have been linked to altered immune function and increased risk for asthma in humans [183, 185].

6.2. Environmental epigenetic modifications

Epigenetic modifications, independent of imprinted genes, can be induced throughout life via interaction with the environment. One of the most well studied examples of epigenetic modifications induced by social environment is the effect of pre-weaning social environment on adult brain and behavior. Post-natal licking and grooming behavior in rats induces epigenetic changes in the glucocorticoid receptor promoter in the hippocampus and in estrogen receptor alpha in the medial preoptic area of the hypothalamus of the offspring that alters HPA axis reactivity which persists into adulthood [59, 294, 325].

The immune system is also amenable to epigenetic modification by environmental factors. Bacterial infection in the bovine udder will inhibit lactation, independent of circulating prolactin levels, via an epigenetic mechanism [285]. Stimulation of human CD4+ T cells induces IL2 expression and epigenetic modifications of the IL2 promoter enhancer which leads to a more rapid and stronger response after a second stimulation, which the authors argue represents an “epigenetic memory” of the regulatory event [213].

6.3. Photoperiodic effects on the epigenome

Although research on the epigenetics of photoperiodism is scant, environmental interactions with the epigenome have been implicated in many of the same social behaviors, such as depression, anxiety, and aggression (reviewed in [65]), that are altered by photoperiod (see previous discussion). More direct evidence of photoperiodic effects on the epigenome come from plants where photoperiodic variations in methylation patterns of genes involved in flowering in Arabidopsis have been identified [73, 278]. Direct evidence for the role of DNA methylation in the control of the photoperiodic flowering response has been reported using Perilla frutescens and Silene armeria, two species of plants with a photoperiod-stable flowering state. The photoperiodic inhibition of flowering in these two species can be reversed using 5-azacytidine, a DNA demethylating agent [165].

Although no direct evidence of photoperiodic effects on the epigenome has been identified in mammals, it has been hypothesized that melatonin, or its metabolites, can function as inhibitors of DNA methyltransferases due to structural similarity with known methyltransferase inhibitors, and thus may be involved in epigenetic regulation [167]. Recent work identifying the specific role of melatonin in epigenetic regulation of several types of cancer has begun to support this theory [168, 215]. Indeed, melatonin plays an important role in cell differentiation during development via interaction with nuclear melatonin receptors, which has been hypothesized to potentially contribute to epigenetic modifications of oocytes to explain adaptive geographic variations in humans [149]. How epigenetic modifications of other genes may be regulated by melatonin, and the potential role in photoperiodism of these genes, remains to be explored.

7. Conclusions

The putative mechanism central to the evolution of photoperiodism in animals is the distribution of energetically challenging activities across the year to optimize reproductive fitness while balancing the energetic tradeoffs necessary for seasonally-appropriate survival strategies (Figure 3). For long-day breeding mammals, the costs of territorial behavior, pregnancy, and lactation constrain reproduction to coincide with the onset of long days when environmental conditions are most conducive to survival. For short-day breeding mammals, lactation appears to be the major energetic cost associated with reproduction; thus, breeding has evolved to occur in fall. Gestation continues throughout the winter and birth and lactation occur in the lengthening days of spring, when food and weather conditions are most conducive to survival. Thus, for both long- and short-day breeders, the energetic costs of supporting reproduction and lactation outweigh the benefits of the investment during times of low reproductive success (viz., winter). Being able to accurately predict future events to allocate energy among competing physiological systems to maximize fitness and survival necessarily requires endogenous mechanisms to permit physiological anticipation of annual conditions. Day length provides the most noise free environmental signal for organisms to monitor and accurately predict time of the year.

Figure 3.

Figure 3

Figure 3

Hypothetical illustration of photoperiodic differences in allocation of energy among competing processes. During the long days of summer, animals are in positive energy balance due to increased availability of resources (food). Thus, energy is available to support somatic growth and reproduction. During the short days of winter, energy balance tips to negative. Reproduction and somatic growth processes are curtailed, and available energy is allocated differentially among the immune system, thermoregulation, and cellular metabolism. As stressors mount immune function is eventually compromised. Often animals in the field display compromised immunity and health during the winter. Presumably, compromised immune function would be worse in the absence of short-day bolstering [224]. Elevated energetic constraints and thermoregulatory demands in the short days of winter have led to the evolution of torpor and hibernation to promote survival by further reduction in the energetic demands of the remaining active processes.

Day length is an unambiguous environmental signal. This signal can be used in the lab to explore the mechanisms by which the environment and genes interact to impart phenotype. Photoperiod manipulation is advantageous over other accepted methods of studying gene – environment interactions, where differential responses to “environment” (which entail an unlimited number of variables) are identified, and then comparative genomic methods are used to identify underlying genetic susceptibility factors (see http://www.niehs.nih.gov/). This unique approach allows isolation of a single environmental factor (duration of light) that can be controlled in a laboratory setting, and investigation of how this one factor interacts with the genome to affect multiple behavioral, cognitive, and physiological processes across diverse species. Using non-traditional animal models to investigate how photoperiodic modulation of physiological and behavioral systems interacts to impart phenotype may contribute to our understanding of clinical disorders and other pathologies in a translational setting.

In mammals, melatonin is the hormonal signal of day length. Although acute administration of melatonin can directly impact aspects of physiology and immune function, altering the duration of pineal melatonin secretion drives enduring changes in many physiological systems, including the HPA axis, the HPG axis, the brain-gut axis, the autonomic nervous system, and the immune system. This plastic and complex interplay among these systems is intimately regulated at all levels by melatonin. Thus, melatonin is the fulcrum mediating redistribution of energetic investment among physiological processes to maximize fitness and survival. Whereas many advances have been made in the past several decades, identification of the diversity of systems and multimodal sites of action of melatonin in photoperiodic animals is an ongoing process. Much of the progress in biology during this time has focused on molecular biology, it is now important to use modern molecular approaches and precise environmental factors to study the development of phenotype. Thus, continued research in the mechanisms of how light interacts with genes via melatonin hormonal signaling can provide novel and important insights into conserved molecular mechanistic themes underlying the relationship between phenotype and genotype.

Acknowledgments

We thank Tracy Bedrosian and Laura Fonken for their helpful comments on this manuscript. Research presented in this manuscript was supported by National Science Foundation Integrative Organismal Systems Grants 0838098 and 0416897, and by National Institute of Mental Health Grant 7R01MH057535-19 to RJN.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Adam CL, Mercer JG. Appetite regulation and seasonality: implications for obesity. Proc Nutr Soc. 2004;63:413–419. doi: 10.1079/pns2004367. [DOI] [PubMed] [Google Scholar]
  • 2.Albers HE, Rowland CM, Ferris CF. Arginine-vasopressin immunoreactivity is not altered by photoperiod or gonadal hormones in the Syrian hamster (Mesocricetus auratus) Brain Res. 1991;539:137–142. doi: 10.1016/0006-8993(91)90696-s. [DOI] [PubMed] [Google Scholar]
  • 3.Ames A, 3rd, Li YY, Heher EC, Kimble CR. Energy metabolism of rabbit retina as related to function: high cost of Na+ transport. J Neurosci. 1992;12:840–853. doi: 10.1523/JNEUROSCI.12-03-00840.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Andrews RV, Belknap RW. Season affects tolerance of cohabitation by deer mice. Physiol Behav. 1993;53:617–620. doi: 10.1016/0031-9384(93)90163-a. [DOI] [PubMed] [Google Scholar]
  • 5.Andrews RV, Phillips D, Makihara D. Metabolic and thermoregulatory consequences of social behaviors between Microtus townsendii. Comp Biochem Physiol A. 1987;87:345–348. doi: 10.1016/0300-9629(87)90133-2. [DOI] [PubMed] [Google Scholar]
  • 6.Ashkenazy-Frolinger T, Kronfeld-Schor N, Juetten J, Einat H. It is darkness and not light: Depression-like behaviors of diurnal unstriped Nile grass rats maintained under a short photoperiod schedule. J Neurosci Methods. 2010;186:165–170. doi: 10.1016/j.jneumeth.2009.11.013. [DOI] [PubMed] [Google Scholar]
  • 7.Ashkenazy T, Einat H, Kronfeld-Schor N. Effects of bright light treatment on depression- and anxiety-like behaviors of diurnal rodents maintained on a short daylight schedule. Behav Brain Res. 2009;201:343–346. doi: 10.1016/j.bbr.2009.03.005. [DOI] [PubMed] [Google Scholar]
  • 8.Ashkenazy T, Einat H, Kronfeld-Schor N. We are in the dark here: induction of depression- and anxiety-like behaviours in the diurnal fat sand rat, by short daylight or melatonin injections. Int J Neuropsychopharmacol. 2009;12:83–93. doi: 10.1017/S1461145708009115. [DOI] [PubMed] [Google Scholar]
  • 9.Axelrod J, Wurtman RJ, Snyder SH. Control of Hydroxyindole O-Methyltransferase Activity in the Rat Pineal Gland by Environmental Lighting. J Biol Chem. 1965;240:949–954. [PubMed] [Google Scholar]
  • 10.Baba K, Pozdeyev N, Mazzoni F, Contreras-Alcantara S, Liu C, Kasamatsu M, Martinez-Merlos T, Strettoi E, Iuvone PM, Tosini G. Melatonin modulates visual function and cell viability in the mouse retina via the MT1 melatonin receptor. Proc Natl Acad Sci U S A. 2009;106:15043–15048. doi: 10.1073/pnas.0904400106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101:15718–15723. doi: 10.1073/pnas.0407076101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Backhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A. 2007;104:979–984. doi: 10.1073/pnas.0605374104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Badura LL, Goldman BD. Seasonal regulation of neuroendocrine activity in male Turkish hamsters (Mesocricetus brandti): role of the hypothalamic paraventricular nucleus. Neuroendocrinology. 1992;55:477–484. doi: 10.1159/000126159. [DOI] [PubMed] [Google Scholar]
  • 14.Badura LL, Nunez AA. Photoperiodic modulation of sexual and aggressive behavior in female golden hamsters (Mesocricetus auratus): role of the pineal gland. Horm Behav. 1989;23:27–42. doi: 10.1016/0018-506x(89)90072-x. [DOI] [PubMed] [Google Scholar]
  • 15.Bae HH, Mangels RA, Cho BS, Dark J, Yellon SM, Zucker I. Ventromedial hypothalamic mediation of photoperiodic gonadal responses in male Syrian hamsters. J Biol Rhythms. 1999;14:391–401. doi: 10.1177/074873099129000795. [DOI] [PubMed] [Google Scholar]
  • 16.Bai A, Guo Y, Shen Y, Xie Y, Zhu X, Lu N. Seasonality in flares and months of births of patients with ulcerative colitis in a Chinese population. Dig Dis Sci. 2009;54:1094–1098. doi: 10.1007/s10620-008-0453-1. [DOI] [PubMed] [Google Scholar]
  • 17.Bailey MT, Walton JC, Dowd SE, Weil ZM, Nelson RJ. Photoperiod modulates gut bacteria composition in male Siberian hamsters (Phodopus sungorus) Brain Behav Immun. 2010;24:577–584. doi: 10.1016/j.bbi.2009.12.010. [DOI] [PubMed] [Google Scholar]
  • 18.Barrett P, Ebling FJ, Schuhler S, Wilson D, Ross AW, Warner A, Jethwa P, Boelen A, Visser TJ, Ozanne DM, Archer ZA, Mercer JG, Morgan PJ. Hypothalamic thyroid hormone catabolism acts as a gatekeeper for the seasonal control of body weight and reproduction. Endocrinology. 2007;148:3608–3617. doi: 10.1210/en.2007-0316. [DOI] [PubMed] [Google Scholar]
  • 19.Bartness TJ, Demas GE, Song CK. Seasonal changes in adiposity: the roles of the photoperiod, melatonin and other hormones, and sympathetic nervous system. Exp Biol Med. 2002;227:363–376. doi: 10.1177/153537020222700601. [DOI] [PubMed] [Google Scholar]
  • 20.Bartness TJ, Powers JB, Hastings MH, Bittman EL, Goldman BD. The timed infusion paradigm for melatonin delivery: what has it taught us about the melatonin signal, its reception, and the photoperiodic control of seasonal responses? J Pineal Res. 1993;15:161–190. doi: 10.1111/j.1600-079x.1993.tb00903.x. [DOI] [PubMed] [Google Scholar]
  • 21.Bartness TJ, Song CK. Brain-adipose tissue neural crosstalk. Physiol Behav. 2007;91:343–351. doi: 10.1016/j.physbeh.2007.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bartness TJ, Wade GN. Photoperiodic control of body weight and energy metabolism in Syrian hamsters (Mesocricetus auratus): role of pineal gland, melatonin, gonads, and diet. Endocrinology. 1984;114:492–498. doi: 10.1210/endo-114-2-492. [DOI] [PubMed] [Google Scholar]
  • 23.Basta NO, James PW, Craft AW, McNally RJ. Season of birth and diagnosis for childhood cancer in Northern England, 1968-2005. Paediatr Perinat Epidemiol. 2010;24:309–318. doi: 10.1111/j.1365-3016.2010.01112.x. [DOI] [PubMed] [Google Scholar]
  • 24.Bayes HK, Weir CJ, O'Leary C. Timing of birth and risk of multiple sclerosis in the Scottish population. Eur Neurol. 2010;63:36–40. doi: 10.1159/000268163. [DOI] [PubMed] [Google Scholar]
  • 25.Becker-Andre M, Wiesenberg I, Schaeren-Wiemers N, Andre E, Missbach M, Saurat JH, Carlberg C. Pineal gland hormone melatonin binds and activates an orphan of the nuclear receptor superfamily. J Biol Chem. 1994;269:28531–28534. [PubMed] [Google Scholar]
  • 26.Beery AK, Loo TJ, Zucker I. Day length and estradiol affect same-sex affiliative behavior in the female meadow vole. Horm Behav. 2008;54:153–159. doi: 10.1016/j.yhbeh.2008.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Beery AK, Paul MJ, Routman DM, Zucker I. Maternal photoperiodic history affects offspring development in Syrian hamsters. J Biol Rhythms. 2008;23:445–455. doi: 10.1177/0748730408322985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Beery AK, Zucker I. Oxytocin and same-sex social behavior in female meadow voles. Neuroscience. 2010;169:665–673. doi: 10.1016/j.neuroscience.2010.05.023. [DOI] [PubMed] [Google Scholar]
  • 29.Benabid N, Mesfioui A, Ouichou A. Effects of photoperiod regimen on emotional behaviour in two tests for anxiolytic activity in Wistar rat. Brain Res Bull. 2008;75:53–59. doi: 10.1016/j.brainresbull.2007.07.016. [DOI] [PubMed] [Google Scholar]
  • 30.Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational definition of epigenetics. Genes Dev. 2009;23:781–783. doi: 10.1101/gad.1787609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bester-Meredith JK, Young LJ, Marler CA. Species differences in paternal behavior and aggression in peromyscus and their associations with vasopressin immunoreactivity and receptors. Horm Behav. 1999;36:25–38. doi: 10.1006/hbeh.1999.1522. [DOI] [PubMed] [Google Scholar]
  • 32.Bilbo SD, Dhabhar FS, Viswanathan K, Saul A, Yellon SM, Nelson RJ. Short day lengths augment stress-induced leukocyte trafficking and stress-induced enhancement of skin immune function. Proc Natl Acad Sci U S A. 2002;99:4067–4072. doi: 10.1073/pnas.062001899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bilbo SD, Drazen DL, Quan N, He L, Nelson RJ. Short day lengths attenuate the symptoms of infection in Siberian hamsters. Proc R Soc Lond B Biol Sci. 2002;269:447–454. doi: 10.1098/rspb.2001.1915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bilbo SD, Nelson RJ. Melatonin regulates energy balance and attenuates fever in Siberian hamsters. Endocrinology. 2002;143:2527. doi: 10.1210/endo.143.7.8922. [DOI] [PubMed] [Google Scholar]
  • 35.Bilbo SD, Nelson RJ. Sex differences in photoperiodic and stress-induced enhancement of immune function in Siberian hamsters. Brain Behav Immun. 2003;17:462–472. doi: 10.1016/s0889-1591(03)00063-1. [DOI] [PubMed] [Google Scholar]
  • 36.Bittman EL, Hegarty CM, Layden MQ, Jonassen JA. Influences of photoperiod on sexual behaviour, neuroendocrine steroid receptors and adenohypophysial hormone secretion and gene expression in female golden hamsters. J Mol Endocrinol. 1990;5:15–25. doi: 10.1677/jme.0.0050015. [DOI] [PubMed] [Google Scholar]
  • 37.Bittman EL, Jetton AE, Villalba C, Devries GJ. Effects of photoperiod and androgen on pituitary function and neuropeptide staining in Siberian hamsters. Am J Physiol. 1996;271:R64–72. doi: 10.1152/ajpregu.1996.271.1.R64. [DOI] [PubMed] [Google Scholar]
  • 38.Bittman EL, Karsch FJ. Nightly duration of pineal melatonin secretion determines the reproductive response to inhibitory day length in the ewe. Biol Reprod. 1984;30:585–593. doi: 10.1095/biolreprod30.3.585. [DOI] [PubMed] [Google Scholar]
  • 39.Bittman EL, Kaynard AH, Olster DH, Robinson JE, Yellon SM, Karsch FJ. Pineal melatonin mediates photoperiodic control of pulsatile luteinizing hormone secretion in the ewe. Neuroendocrinology. 1985;40:409–418. doi: 10.1159/000124106. [DOI] [PubMed] [Google Scholar]
  • 40.Bittman EL, Zucker I. Influences of the adrenal gland and photoperiod on the hamster oestrus cycle. J Reprod Fertil. 1977;50:331–333. doi: 10.1530/jrf.0.0500331. [DOI] [PubMed] [Google Scholar]
  • 41.Blank JL, Freeman DA. Differential reproductive response to short photoperiod in deer mice: role of melatonin. J Comp Physiol [A] 1991;169:501–506. doi: 10.1007/BF00197662. [DOI] [PubMed] [Google Scholar]
  • 42.Bliss SP, Navratil AM, Xie J, Roberson MS. GnRH signaling, the gonadotrope and endocrine control of fertility. Front Neuroendocrinol. 2010;31:322–340. doi: 10.1016/j.yfrne.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the hippocampus. Nature. 1993;361:31–39. doi: 10.1038/361031a0. [DOI] [PubMed] [Google Scholar]
  • 44.Blom JM, Gerber JM, Nelson RJ. Day length affects immune cell numbers in deer mice: interactions with age, sex, and prenatal photoperiod. Am J Physiol. 1994;267:R596–601. doi: 10.1152/ajpregu.1994.267.2.R596. [DOI] [PubMed] [Google Scholar]
  • 45.Bonneaud C, Mazuc J, Gonzalez G, Haussy C, Chastel O, Faivre B, Sorci G. Assessing the cost of mounting an immune response. Am Nat. 2003;161:367–379. doi: 10.1086/346134. [DOI] [PubMed] [Google Scholar]
  • 46.Boonstra R. Life history variation in maturation in fluctuating meadow vole populations (Microtus pennsylvanicus) Oikos. 1989;54:265–274. [Google Scholar]
  • 47.Bosshardt D, Ajdacic-Gross V, Lang P, Bosshardt M, Bopp M, Addor MC, Gutzwiller F. Season of birth in valvular heart disease. Paediatr Perinat Epidemiol. 2005;19:246–252. doi: 10.1111/j.1365-3016.2005.00645.x. [DOI] [PubMed] [Google Scholar]
  • 48.Bowers RR, Gettys TW, Prpic V, Harris RB, Bartness TJ. Short photoperiod exposure increases adipocyte sensitivity to noradrenergic stimulation in Siberian hamsters. Am J Physiol Regul Integr Comp Physiol. 2005;288:R1354–1360. doi: 10.1152/ajpregu.00792.2004. [DOI] [PubMed] [Google Scholar]
  • 49.Bradshaw WE, Holzapfel CM. Genetic shift in photoperiodic response correlated with global warming. Proc Natl Acad Sci U S A. 2001;98:14509–14511. doi: 10.1073/pnas.241391498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bronson FH. Mammalian reproduction: an ecological perspective. Biol Reprod. 1985;32:1–26. doi: 10.1095/biolreprod32.1.1. [DOI] [PubMed] [Google Scholar]
  • 51.Butler MP, Turner KW, Park JH, Schoomer EE, Zucker I, Gorman MR. Seasonal regulation of reproduction: altered role of melatonin under naturalistic conditions in hamsters. Proc Biol Sci. 2010;277:2867–2874. doi: 10.1098/rspb.2010.0396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Caldwell GS, Glickman SE, Smith ER. Seasonal aggression independent of seasonal testosterone in wood rats. Proc Natl Acad Sci U S A. 1984;81:5255–5257. doi: 10.1073/pnas.81.16.5255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Caldwell HK, Albers HE. Short-photoperiod exposure reduces vasopressin (V1a) receptor binding but not arginine-vasopressin-induced flank marking in male Syrian hamsters. J Neuroendocrinol. 2003;15:971–977. doi: 10.1046/j.1365-2826.2003.01086.x. [DOI] [PubMed] [Google Scholar]
  • 54.Campbell CS, Tabor J, Davis JD. Small effect of brown adipose tissue and major effect of photoperiod on body weight in hamsters (Mesocricetus auratus) Physiol Behav. 1983;30:349–352. doi: 10.1016/0031-9384(83)90137-3. [DOI] [PubMed] [Google Scholar]
  • 55.Card JP, Whealy ME, Robbins AK, Moore RY, Enquist LW. Two alphaherpesvirus strains are transported differentially in the rodent visual system. Neuron. 1991;6:957–969. doi: 10.1016/0896-6273(91)90236-s. [DOI] [PubMed] [Google Scholar]
  • 56.Carter DS, Goldman BD. Antigonadal effects of timed melatonin infusion in pinealectomized male Djungarian hamsters (Phodopus sungorus sungorus): duration is the critical parameter. Endocrinology. 1983;113:1261–1267. doi: 10.1210/endo-113-4-1261. [DOI] [PubMed] [Google Scholar]
  • 57.Ceesay SM, Prentice AM, Cole TJ, Foord F, Weaver LT, Poskitt EM, Whitehead RG. Effects on birth weight and perinatal mortality of maternal dietary supplements in rural Gambia: 5 year randomised controlled trial. BMJ. 1997;315:786–790. doi: 10.1136/bmj.315.7111.786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chalivoix S, Malpaux B, Dufourny L. Relationship between polysialylated neural cell adhesion molecule and beta-endorphin- or gonadotropin releasing hormone-containing neurons during activation of the gonadotrope axis in short daylength in the ewe. Neuroscience. 2010;169:1326–1336. doi: 10.1016/j.neuroscience.2010.05.045. [DOI] [PubMed] [Google Scholar]
  • 59.Champagne FA, Weaver IC, Diorio J, Sharma S, Meaney MJ. Natural variations in maternal care are associated with estrogen receptor alpha expression and estrogen sensitivity in the medial preoptic area. Endocrinology. 2003;144:4720–4724. doi: 10.1210/en.2003-0564. [DOI] [PubMed] [Google Scholar]
  • 60.Chattoraj A, Liu T, Zhang LS, Huang Z, Borjigin J. Melatonin formation in mammals: in vivo perspectives. Rev Endocr Metab Disord. 2009;10:237–243. doi: 10.1007/s11154-009-9125-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Choudhuri S, Cui Y, Klaassen CD. Molecular targets of epigenetic regulation and effectors of environmental influences. Toxicol Appl Pharmacol. 2010;245:378–393. doi: 10.1016/j.taap.2010.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chowdhury VS, Yamamoto K, Ubuka T, Bentley GE, Hattori A, Tsutsui K. Melatonin stimulates the release of gonadotropin-inhibitory hormone by the avian hypothalamus. Endocrinology. 2010;151:271–280. doi: 10.1210/en.2009-0908. [DOI] [PubMed] [Google Scholar]
  • 63.Clarke MB, Sperandio V. Events at the host-microbial interface of the gastrointestinal tract III. Cell-to-cell signaling among microbial flora, host, and pathogens: there is a whole lot of talking going on. Am J Physiol Gastrointest Liver Physiol. 2005;288:G1105–1109. doi: 10.1152/ajpgi.00572.2004. [DOI] [PubMed] [Google Scholar]
  • 64.Collinson AC, Moore SE, Cole TJ, Prentice AM. Birth season and environmental influences on patterns of thymic growth in rural Gambian infants. Acta Paediatr. 2003;92:1014–1020. [PubMed] [Google Scholar]
  • 65.Crews D. Epigenetics, brain, behavior, and the environment. Hormones. 2010;9:41–50. doi: 10.14310/horm.2002.1251. [DOI] [PubMed] [Google Scholar]
  • 66.Csaba G, Barath P. Morphological changes of thymus and the thyroid gland after postnatal extirpation of pineal body. Endocrinol Exp. 1975;9:59–67. [PubMed] [Google Scholar]
  • 67.Dark J, Dark KA, Zucker I. Long day lengths increase brain weight and DNA content in the meadow vole, Microtus pennsylvanicus. Brain Res. 1987;409:302–307. doi: 10.1016/0006-8993(87)90715-3. [DOI] [PubMed] [Google Scholar]
  • 68.Dark J, Zucker I. Gonadal and photoperiodic control of seasonal body weight changes in male voles. Am J Physiol. 1984;247:R84–88. doi: 10.1152/ajpregu.1984.247.1.R84. [DOI] [PubMed] [Google Scholar]
  • 69.Dark J, Zucker I. Seasonal cycles in energy balance: regulation by light. Ann N Y Acad Sci. 1985;453:170–181. doi: 10.1111/j.1749-6632.1985.tb11809.x. [DOI] [PubMed] [Google Scholar]
  • 70.Dark J, Zucker I, Wade GN. Photoperiodic regulation of body mass, food intake, and reproduction in meadow voles. Am J Physiol. 1983;245:R334–338. doi: 10.1152/ajpregu.1983.245.3.R334. [DOI] [PubMed] [Google Scholar]
  • 71.Davies W, Lynn PM, Relkovic D, Wilkinson LS. Imprinted genes and neuroendocrine function. Front Neuroendocrinol. 2008;29:413–427. doi: 10.1016/j.yfrne.2007.12.001. [DOI] [PubMed] [Google Scholar]
  • 72.Dehnel A. Studies on the genus Sorex L. Ann Univ Mariae Curie Sklodowska. 1949;4:17–97. [Google Scholar]
  • 73.del Olmo I, Lopez-Gonzalez L, Martin-Trillo MM, Martinez-Zapater JM, Pineiro M, Jarillo JA. EARLY IN SHORT DAYS 7 (ESD7) encodes the catalytic subunit of DNA polymerase epsilon and is required for flowering repression through a mechanism involving epigenetic gene silencing. Plant J. 2010;61:623–636. doi: 10.1111/j.1365-313X.2009.04093.x. [DOI] [PubMed] [Google Scholar]
  • 74.Delagrange P, Atkinson J, Boutin JA, Casteilla L, Lesieur D, Misslin R, Pellissier S, Penicaud L, Renard P. Therapeutic perspectives for melatonin agonists and antagonists. J Neuroendocrinol. 2003;15:442–448. doi: 10.1046/j.1365-2826.2003.01016.x. [DOI] [PubMed] [Google Scholar]
  • 75.Demas G. In vivo but not in vitro leptin enhances lymphocyte proliferation in Siberian hamsters (Phodopus sungorus) Gen Comp Endocrinol. 2009 doi: 10.1016/j.ygcen.2009.10.011. [DOI] [PubMed] [Google Scholar]
  • 76.Demas GE. Splenic denervation blocks leptin-induced enhancement of humoral immunity in Siberian hamsters (Phodopus sungorus) Neuroendocrinology. 2002;76:178–184. doi: 10.1159/000064527. [DOI] [PubMed] [Google Scholar]
  • 77.Demas GE. The energetics of immunity: a neuroendocrine link between energy balance and immune function. Horm Behav. 2004;45:173–180. doi: 10.1016/j.yhbeh.2003.11.002. [DOI] [PubMed] [Google Scholar]
  • 78.Demas GE, Bartness TJ, Nelson RJ, Drazen DL. Photoperiod modulates the effects of norepinephrine on lymphocyte proliferation in Siberian hamsters. Am J Physiol. 2003;285:R873–879. doi: 10.1152/ajpregu.00209.2003. [DOI] [PubMed] [Google Scholar]
  • 79.Demas GE, Chefer V, Talan MI, Nelson RJ. Metabolic costs of mounting an antigen-stimulated immune response in adult and aged C57BL/6J mice. Am J Physiol. 1997;273:R1631–1637. doi: 10.1152/ajpregu.1997.273.5.R1631. [DOI] [PubMed] [Google Scholar]
  • 80.Demas GE, DeVries AC, Nelson RJ. Effects of photoperiod and 2-deoxy-D-glucose-induced metabolic stress on immune function in female deer mice. Am J Physiol. 1997;272:R1762–1767. doi: 10.1152/ajpregu.1997.272.6.R1762. [DOI] [PubMed] [Google Scholar]
  • 81.Demas GE, Drazen DL, Jasnow AM, Bartness TJ, Nelson RJ. Sympathoadrenal system differentially affects photoperiodic changes in humoral immunity of Siberian hamsters (Phodopus sungorus) J Neuroendocrinol. 2002;14:29–35. doi: 10.1046/j.0007-1331.2001.00736.x. [DOI] [PubMed] [Google Scholar]
  • 82.Demas GE, Drazen DL, Nelson RJ. Reductions in total body fat decrease humoral immunity. Proc Biol Sci. 2003;270:905–911. doi: 10.1098/rspb.2003.2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Demas GE, Nelson RJ. Photoperiod and temperature interact to affect immune parameters in adult male deer mice (Peromyscus maniculatus) J Biol Rhythms. 1996;11:94–102. doi: 10.1177/074873049601100202. [DOI] [PubMed] [Google Scholar]
  • 84.Demas GE, Nelson RJ. Exogenous melatonin enhances cell-mediated, but not humoral, immune function in adult male deer mice (Peromyscus maniculatus) J Biol Rhythms. 1998;13:245–252. doi: 10.1177/074873098129000084. [DOI] [PubMed] [Google Scholar]
  • 85.Demas GE, Nelson RJ. Photoperiod, ambient temperature, and food availability interact to affect reproductive and immune function in adult male deer mice. J Biol Rhythms. 1998;13:253. doi: 10.1177/074873098129000093. [DOI] [PubMed] [Google Scholar]
  • 86.Demas GE, Nelson RJ. Short-day enhancement of immune function is independent of steroid hormones in deer mice (Peromyscus maniculatus) J Comp Physiol [B] 1998;168:419–426. doi: 10.1007/s003600050161. [DOI] [PubMed] [Google Scholar]
  • 87.Demas GE, Polacek KM, Durazzo A, Jasnow AM. Adrenal hormones mediate melatonin-induced increases in aggression in male Siberian hamsters (Phodopus sungorus) Horm Behav. 2004;46:582–591. doi: 10.1016/j.yhbeh.2004.07.001. [DOI] [PubMed] [Google Scholar]
  • 88.Demas GE, Sakaria S. Leptin regulates energetic tradeoffs between body fat and humoural immunity. Proc Biol Sci. 2005;272:1845–1850. doi: 10.1098/rspb.2005.3126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Dome P, Kapitany B, Ignits G, Rihmer Z. Season of birth is significantly associated with the risk of completed suicide. Biol Psychiatry. 2010;68:148–155. doi: 10.1016/j.biopsych.2010.03.005. [DOI] [PubMed] [Google Scholar]
  • 90.Drazen DL, Demas GE, Nelson RJ. Leptin effects on immune function and energy balance are photoperiod dependent in Siberian hamsters (Phodopus sungorus) Endocrinology. 2001;142:2768–2775. doi: 10.1210/endo.142.7.8271. [DOI] [PubMed] [Google Scholar]
  • 91.Dubocovich ML. Melatonin is a potent modulator of dopamine release in the retina. Nature. 1983;306:782–784. doi: 10.1038/306782a0. [DOI] [PubMed] [Google Scholar]
  • 92.Dubocovich ML. Melatonin receptors: are there multiple subtypes? Trends Pharmacol Sci. 1995;16:50–56. doi: 10.1016/s0165-6147(00)88978-6. [DOI] [PubMed] [Google Scholar]
  • 93.Dubocovich ML, Markowska M. Functional MT1 and MT2 melatonin receptors in mammals. Endocrine. 2005;27:101–110. doi: 10.1385/ENDO:27:2:101. [DOI] [PubMed] [Google Scholar]
  • 94.Dubocovich ML, Rivera-Bermudez MA, Gerdin MJ, Masana MI. Molecular pharmacology, regulation and function of mammalian melatonin receptors. Front Biosci. 2003;8:d1093–1108. doi: 10.2741/1089. [DOI] [PubMed] [Google Scholar]
  • 95.Dulac C. Brain function and chromatin plasticity. Nature. 2010;465:728–735. doi: 10.1038/nature09231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Duncan MJ, Goldman BD, Di Pinto MN, Stetson MH. Testicular function and pelage color have different critical daylengths in the Djungarian hamster, Phodopus sungorus sungorus. Endocrinology. 1985;116:424–430. doi: 10.1210/endo-116-1-424. [DOI] [PubMed] [Google Scholar]
  • 97.Ebisawa T, Karne S, Lerner MR, Reppert SM. Expression cloning of a high-affinity melatonin receptor from Xenopus dermal melanophores. Proc Natl Acad Sci U S A. 1994;91:6133–6137. doi: 10.1073/pnas.91.13.6133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Elliot JA. Circadian rhythms, entrainment and photoperiodism in the Syrian hamster. In: Follett BK, Follett DE, editors. Biological Clocks in Seasonal Reproductive Cycles. Scientechnica; Bristol: 1981. pp. 203–217. [Google Scholar]
  • 99.Ergon T, MacKinnon JL, Stenseth N, Boonstra R, Lambin X. Mechanisms for Delayed Density-Dependent Reproductive Traits in Field Voles, Microtus agrestis: The Importance of Inherited Environmental Effects. Oikos. 2001;95:185–197. [Google Scholar]
  • 100.Falcon J, Besseau L, Fuentes M, Sauzet S, Magnanou E, Boeuf G. Structural and functional evolution of the pineal melatonin system in vertebrates. Ann N Y Acad Sci. 2009;1163:101–111. doi: 10.1111/j.1749-6632.2009.04435.x. [DOI] [PubMed] [Google Scholar]
  • 101.Ferris CF. Vasopressin/oxytocin and aggression. Novartis Found Symp. 2005;268:190–198. discussion 198-200, 242-153. [PubMed] [Google Scholar]
  • 102.Fine JB, Bartness TJ. Daylength and body mass affect diet self-selection by Siberian hamsters. Physiol Behav. 1996;59:1039–1050. doi: 10.1016/0031-9384(95)02240-6. [DOI] [PubMed] [Google Scholar]
  • 103.Fleming AS, Phillips A, Rydall A, Levesque L. Effects of photoperiod, the pineal gland and the gonads on agonistic behavior in female golden hamsters (Mesocricetus auratus) Physiol Behav. 1988;44:227–234. doi: 10.1016/0031-9384(88)90143-6. [DOI] [PubMed] [Google Scholar]
  • 104.Foster RG, Roenneberg T. Human responses to the geophysical daily, annual and lunar cycles. Curr Biol. 2008;18:R784–R794. doi: 10.1016/j.cub.2008.07.003. [DOI] [PubMed] [Google Scholar]
  • 105.Fountoulakis KN, Iacovides A, Karamouzis M, Kaprinis GS, Ierodiakonou C. Season of birth, clinical manifestations and Dexamethasone Suppression Test in unipolar major depression. Ann Gen Psychiatry. 2007;6:20. doi: 10.1186/1744-859X-6-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Frangos E, Athanassenas G, Tsitourides S, Psilolignos P, Robos A, Katsanou N, Bulgaris C. Seasonality of the episodes of recurrent affective psychoses. Possible prophylactic interventions. J Affect Disord. 1980;2:239–247. doi: 10.1016/0165-0327(80)90025-7. [DOI] [PubMed] [Google Scholar]
  • 107.Freeman DA, Teubner BJ, Smith CD, Prendergast BJ. Exogenous T3 mimics long day lengths in Siberian hamsters. Am J Physiol Regul Integr Comp Physiol. 2007;292:R2368–2372. doi: 10.1152/ajpregu.00713.2006. [DOI] [PubMed] [Google Scholar]
  • 108.Frost D, Zucker I. Photoperiod and melatonin influence seasonal gonadal cycles in the grasshopper mouse (Onychomys leucogaster) J Reprod Fertil. 1983;69:237–244. doi: 10.1530/jrf.0.0690237. [DOI] [PubMed] [Google Scholar]
  • 109.Galea LA, Kavaliers M, Ossenkopp KP. Sexually dimorphic spatial learning in meadow voles Microtus pennsylvanicus and deer mice Peromyscus maniculatus. J Exp Biol. 1996;199:195–200. doi: 10.1242/jeb.199.1.195. [DOI] [PubMed] [Google Scholar]
  • 110.Galea LA, Kavaliers M, Ossenkopp KP, Innes D, Hargreaves EL. Sexually dimorphic spatial learning varies seasonally in two populations of deer mice. Brain Res. 1994;635:18–26. doi: 10.1016/0006-8993(94)91419-2. [DOI] [PubMed] [Google Scholar]
  • 111.Galea LA, McEwen BS. Sex and seasonal differences in the rate of cell proliferation in the dentate gyrus of adult wild meadow voles. Neuroscience. 1999;89:955–964. doi: 10.1016/s0306-4522(98)00345-5. [DOI] [PubMed] [Google Scholar]
  • 112.Garrett JW, Campbell CS. Changes in social behavior of the male golden hamster accompanying photoperiodic changes in reproduction. Horm Behav. 1980;14:303–318. doi: 10.1016/0018-506x(80)90020-3. [DOI] [PubMed] [Google Scholar]
  • 113.Gingerich S, Wang X, Lee PK, Dhillon SS, Chalmers JA, Koletar MM, Belsham DD. The generation of an array of clonal, immortalized cell models from the rat hypothalamus: analysis of melatonin effects on kisspeptin and gonadotropin-inhibitory hormone neurons. Neuroscience. 2009;162:1134–1140. doi: 10.1016/j.neuroscience.2009.05.026. [DOI] [PubMed] [Google Scholar]
  • 114.Godson C, Reppert SM. The Mel1a melatonin receptor is coupled to parallel signal transduction pathways. Endocrinology. 1997;138:397–404. doi: 10.1210/endo.138.1.4824. [DOI] [PubMed] [Google Scholar]
  • 115.Gold DR, Bloomberg GR, Cruikshank WW, Visness CM, Schwarz J, Kattan M, O'Connor GT, Wood RA, Burger MS, Wright RJ, Witter F, Lee-Parritz A, Sperling R, Sadovsky Y, Togias A, Gern JE. Parental characteristics, somatic fetal growth, and season of birth influence innate and adaptive cord blood cytokine responses. J Allergy Clin Immunol. 2009;124:1078–1087. doi: 10.1016/j.jaci.2009.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Goldman BD. Mammalian photoperiodic system: formal properties and neuroendocrine mechanisms of photoperiodic time measurement. J Biol Rhythms. 2001;16:283–301. doi: 10.1177/074873001129001980. [DOI] [PubMed] [Google Scholar]
  • 117.Goldman BD, Nelson RJ. Melatonin and seasonality in mammals. In: Yu HS, Reiter RJ, editors. Melatonin : biosynthesis, physiological effects, and clinical applications. CRC Press; Boca Raton: 1993. p. 550. [Google Scholar]
  • 118.Gonzalez DA, Victora CG, Goncalves H. The effects of season at time of birth on asthma and pneumonia in childhood and adulthood in a birth cohort in southern Brazil. Cad Saude Publica. 2008;24:1089–1102. doi: 10.1590/s0102-311x2008000500016. [DOI] [PubMed] [Google Scholar]
  • 119.Goodson JL. Nonapeptides and the evolutionary patterning of sociality. Prog Brain Res. 2008;170:3–15. doi: 10.1016/S0079-6123(08)00401-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Gorman MR. A plastic interval timer synchronizes pubertal development of summer- and fall-born hamsters. Am J Physiol Regul Integr Comp Physiol. 2001;281:R1613–1623. doi: 10.1152/ajpregu.2001.281.5.R1613. [DOI] [PubMed] [Google Scholar]
  • 121.Gorman MR, Zucker I. Seasonal adaptations of Siberian hamsters. II. Pattern of change in daylength controls annual testicular and body weight rhythms. Biol Reprod. 1995;53:116–125. doi: 10.1095/biolreprod53.1.116. [DOI] [PubMed] [Google Scholar]
  • 122.Green DA, Millar JS. Changes in gut dimensions and capacity of Peromyscus maniculatus relative to diet quality and energy needs. Can J Zool. 1987;65:2159–2162. [Google Scholar]
  • 123.Greives TJ, Humber SA, Goldstein AN, Scotti MA, Demas GE, Kriegsfeld LJ. Photoperiod and testosterone interact to drive seasonal changes in kisspeptin expression in Siberian hamsters (Phodopus sungorus) J Neuroendocrinol. 2008;20:1339–1347. doi: 10.1111/j.1365-2826.2008.01790.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Greives TJ, Kriegsfeld LJ, Bentley GE, Tsutsui K, Demas GE. Recent advances in reproductive neuroendocrinology: a role for RFamide peptides in seasonal reproduction? Proc Biol Sci. 2008;275:1943–1951. doi: 10.1098/rspb.2008.0433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Greives TJ, Mason AO, Scotti MA, Levine J, Ketterson ED, Kriegsfeld LJ, Demas GE. Environmental control of kisspeptin: implications for seasonal reproduction. Endocrinology. 2007;148:1158–1166. doi: 10.1210/en.2006-1249. [DOI] [PubMed] [Google Scholar]
  • 126.Gross JE, Wang Z, Wunder BA. Effects of food quality and energy needs: changes in gut morphology and capacity of Microtus ochrogaster. J Mammal. 1985;66:661–667. [Google Scholar]
  • 127.Grover V, Lipton RB, Sclove SL. Seasonality of month of birth among African American children with diabetes mellitus in the city of Chicago. J Pediatr Endocrinol Metab. 2004;17:289–296. doi: 10.1515/jpem.2004.17.3.289. [DOI] [PubMed] [Google Scholar]
  • 128.Guerrero JM, Reiter RJ. Melatonin-immune system relationships. Curr Top Med Chem. 2002;2:167–179. doi: 10.2174/1568026023394335. [DOI] [PubMed] [Google Scholar]
  • 129.Gundersen G, Andreassen HP. Causes and consequences of natal dispersal in root voles, Microtus oeconomus. Anim Behav. 1998;56:1355–1366. doi: 10.1006/anbe.1998.0911. [DOI] [PubMed] [Google Scholar]
  • 130.Hadley AR, Tran LT, Fagoaga OR, Nehlsen-Cannarella SL, Yellon SM. Sex differences in photoperiod control of antigen-specific primary and secondary humoral immunity in Siberian hamsters. J Neuroimmunol. 2002;128:39–48. doi: 10.1016/s0165-5728(02)00144-3. [DOI] [PubMed] [Google Scholar]
  • 131.Haldar C, Ahmad R. Photoimmunomodulation and melatonin. J Photochem Photobiol B. 2010;98:107–117. doi: 10.1016/j.jphotobiol.2009.11.014. [DOI] [PubMed] [Google Scholar]
  • 132.Haller J, Kruck MR. Neuroendocrine stress responses and aggression. In: Mattson MP, editor. Neurobiology of aggression: understanding and preventing violencery neuroscience. Humana Press; Totowa, N.J.: 2003. pp. 93–118. [Google Scholar]
  • 133.Hannu T, Jaakkola MS, Kivisaari L, Huuskonen MS, Vehmas T. Season of birth and lung fibrosis among workers exposed to asbestos. Chronobiol Int. 2007;24:539–551. doi: 10.1080/07420520701420725. [DOI] [PubMed] [Google Scholar]
  • 134.Hanon EA, Routledge K, Dardente H, Masson-Pevet M, Morgan PJ, Hazlerigg DG. Effect of photoperiod on the thyroid-stimulating hormone neuroendocrine system in the European hamster (Cricetus cricetus) J Neuroendocrinol. 2010;22:51–55. doi: 10.1111/j.1365-2826.2009.01937.x. [DOI] [PubMed] [Google Scholar]
  • 135.Hare EH. Variations in the seasonal distribution of births of psychotic patients in England and Wales. Br J Psychiatry. 1978;132:155–158. doi: 10.1192/bjp.132.2.155. [DOI] [PubMed] [Google Scholar]
  • 136.Hazlerigg D. Genetic and Molecular Mechanisms of Mammalian Photoperiodism. In: Nelson RJ, Denlinger DL, Somers DE, editors. Photoperiodism: the biological calendar. Oxford University Press; Oxford; New York: 2010. pp. 543–560. [Google Scholar]
  • 137.Hazlerigg D, Loudon A. New insights into ancient seasonal life timers. Curr Biol. 2008;18:R795–R804. doi: 10.1016/j.cub.2008.07.040. [DOI] [PubMed] [Google Scholar]
  • 138.Hazlerigg DG, Wagner GC. Seasonal photoperiodism in vertebrates: from coincidence to amplitude. Trends Endocrinol Metab. 2006;17:83–91. doi: 10.1016/j.tem.2006.02.004. [DOI] [PubMed] [Google Scholar]
  • 139.Heldmaier G, Steinlechner S, Ruf T, Wiesinger H, Klingenspor M. Photoperiod and thermoregulation in vertebrates: body temperature rhythms and thermogenic acclimation. J Biol Rhythms. 1989;4:251–265. [PubMed] [Google Scholar]
  • 140.Helwig M, Archer ZA, Heldmaier G, Tups A, Mercer JG, Klingenspor M. Photoperiodic regulation of satiety mediating neuropeptides in the brainstem of the seasonal Siberian hamster (Phodopus sungorus) J Comp Physiol A Neuroethol Sens Neural Behav Physiol. 2009;195:631–642. doi: 10.1007/s00359-009-0438-3. [DOI] [PubMed] [Google Scholar]
  • 141.Henry BA, Blache D, Dunshea FR, Clarke IJ. Altered “set-point” of the hypothalamus determines effects of cortisol on food intake, adiposity, and metabolic substrates in sheep. Domest Anim Endocrinol. 2010;38:46–56. doi: 10.1016/j.domaniend.2009.07.006. [DOI] [PubMed] [Google Scholar]
  • 142.Hoffman RA. Seasonal growth and development and the influence of the eyes and pineal gland on body weight of golden hamsters (M. auratus) Growth. 1983;47:109–121. [PubMed] [Google Scholar]
  • 143.Hoffman RA, Davidson K, Steinberg K. Influence of photoperiod and temperature on weight gain, food consumption, fat pads and thyroxine in male golden hamsters. Growth. 1982;46:150–162. [PubMed] [Google Scholar]
  • 144.Hoffmann K. Effects of short photoperiods on puberty, growth and moult in the Djungarian hamster (Phodopus sungorus) J Reprod Fertil. 1978;54:29–35. doi: 10.1530/jrf.0.0540029. [DOI] [PubMed] [Google Scholar]
  • 145.Horton TH. Growth and reproductive development of male Microtus montanus is affected by the prenatal photoperiod. Biol Reprod. 1984;31:499–504. doi: 10.1095/biolreprod31.3.499. [DOI] [PubMed] [Google Scholar]
  • 146.Horton TH, Buxton OM, Losee-Olson S, Turek FW. Twenty-four-hour profiles of serum leptin in siberian and golden hamsters: photoperiodic and diurnal variations. Horm Behav. 2000;37:388–398. doi: 10.1006/hbeh.2000.1592. [DOI] [PubMed] [Google Scholar]
  • 147.Hughes DT, Clarke MB, Yamamoto K, Rasko DA, Sperandio V. The QseC adrenergic signaling cascade in Enterohemorrhagic E. coli (EHEC) PLoS Pathog. 2009;5:e1000553. doi: 10.1371/journal.ppat.1000553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Hume D, Beiglbock C, Ruf T, Frey-Roos F, Bruns U, Arnold W. Seasonal changes in morphology and function of the gastrointestinal tract of free-living alpine marmots (Marmota marmota) J Comp Physiol [B] 2002;172:197–207. doi: 10.1007/s00360-001-0240-1. [DOI] [PubMed] [Google Scholar]
  • 149.Irmak MK, Topal T, Oter S. Melatonin seems to be a mediator that transfers the environmental stimuli to oocytes for inheritance of adaptive changes through epigenetic inheritance system. Med Hypotheses. 2005;64:1138–1143. doi: 10.1016/j.mehy.2004.12.014. [DOI] [PubMed] [Google Scholar]
  • 150.Jacobs LF. The economy of winter: phenotypic plasticity in behavior and brain structure. Biol Bull. 1996;191:92–100. doi: 10.2307/1543068. [DOI] [PubMed] [Google Scholar]
  • 151.Jasnow AM, Huhman KL, Bartness TJ, Demas GE. Short-day increases in aggression are inversely related to circulating testosterone concentrations in male Siberian hamsters (Phodopus sungorus) Horm Behav. 2000;38:102–110. doi: 10.1006/hbeh.2000.1604. [DOI] [PubMed] [Google Scholar]
  • 152.Jasnow AM, Huhman KL, Bartness TJ, Demas GE. Short days and exogenous melatonin increase aggression of male Syrian hamsters (Mesocricetus auratus) Horm Behav. 2002;42:13–20. doi: 10.1006/hbeh.2002.1797. [DOI] [PubMed] [Google Scholar]
  • 153.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074–1080. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
  • 154.Jockers R, Maurice P, Boutin JA, Delagrange P. Melatonin receptors, heterodimerization, signal transduction and binding sites: what's new? Br J Pharmacol. 2008;154:1182–1195. doi: 10.1038/bjp.2008.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Johnston JD, Tournier BB, Andersson H, Masson-Pevet M, Lincoln GA, Hazlerigg DG. Multiple effects of melatonin on rhythmic clock gene expression in the mammalian pars tuberalis. Endocrinology. 2006;147:959–965. doi: 10.1210/en.2005-1100. [DOI] [PubMed] [Google Scholar]
  • 156.Juszczak M. The hypothalamo-neurohypophysial response to melatonin. Neuroendocrinol Lett. 2001;22:169–174. [PubMed] [Google Scholar]
  • 157.Kabithe EW, Place NJ. Photoperiod-dependent modulation of anti-Mullerian hormone in female Siberian hamsters, Phodopus sungorus. Reproduction. 2008;135:335–342. doi: 10.1530/REP-07-0423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Karsch FJ, Legan SJ, Ryan KD, Foster DL. Importance of estradiol and progesterone in regulating LH secretion and estrous behavior during the sheep estrous cycle. Biol Reprod. 1980;23:404–413. doi: 10.1095/biolreprod23.2.404. [DOI] [PubMed] [Google Scholar]
  • 159.Kauffman AS, Freeman DA, Zucker I. Termination of neuroendocrine refractoriness to melatonin in Siberian hamsters (Phodopus sungorus) J Neuroendocrinol. 2003;15:191–196. doi: 10.1046/j.1365-2826.2003.00966.x. [DOI] [PubMed] [Google Scholar]
  • 160.Kirkpatrick B, Messias E, LaPorte D. Schizoid-like features and season of birth in a nonpatient sample. Schizophr Res. 2008;103:151–155. doi: 10.1016/j.schres.2007.12.479. [DOI] [PubMed] [Google Scholar]
  • 161.Klasing KC. The costs of immunity. Acta Zoologica Sinica. 2004;50:961–969. [Google Scholar]
  • 162.Klein SL. Sex Differences in Infectious and Autoimmune Diseases. In: Becker JB, Berkley JK, Geary N, Hampson E, Herman JP, Young E, editors. Sex differences in the brain: from genes to behavior. Oxford University Press; USA, New York: 2008. pp. 329–353. [Google Scholar]
  • 163.Knudsen TB, Thomsen SF, Ulrik CS, Fenger M, Nepper-Christensen S, Backer V. Season of birth and risk of atopic disease among children and adolescents. J Asthma. 2007;44:257–260. doi: 10.1080/02770900701246832. [DOI] [PubMed] [Google Scholar]
  • 164.Ko CH, Takahashi JS. Molecular components of the mammalian circadian clock. Hum Mol Genet 15 Spec No. 2006;2:R271–277. doi: 10.1093/hmg/ddl207. [DOI] [PubMed] [Google Scholar]
  • 165.Kondo H, Miura T, Wada KC, Takeno K. Induction of flowering by 5-azacytidine in some plant species: relationship between the stability of photoperiodically induced flowering and flower-inducing effect of DNA demethylation. Physiol Plant. 2007;131:462–469. doi: 10.1111/j.1399-3054.2007.00965.x. [DOI] [PubMed] [Google Scholar]
  • 166.Kong WM, Martin NM, Smith KL, Gardiner JV, Connoley IP, Stephens DA, Dhillo WS, Ghatei MA, Small CJ, Bloom SR. Triiodothyronine stimulates food intake via the hypothalamic ventromedial nucleus independent of changes in energy expenditure. Endocrinology. 2004;145:5252–5258. doi: 10.1210/en.2004-0545. [DOI] [PubMed] [Google Scholar]
  • 167.Korkmaz A, Reiter RJ. Epigenetic regulation: a new research area for melatonin? J Pineal Res. 2008;44:41–44. doi: 10.1111/j.1600-079X.2007.00509.x. [DOI] [PubMed] [Google Scholar]
  • 168.Korkmaz A, Sanchez-Barcelo EJ, Tan DX, Reiter RJ. Role of melatonin in the epigenetic regulation of breast cancer. Breast Cancer Res Treat. 2009;115:13–27. doi: 10.1007/s10549-008-0103-5. [DOI] [PubMed] [Google Scholar]
  • 169.Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, Sar M, Korach KS, Gustafsson JA, Smithies O. Generation and reproductive phenotypes of mice lacking estrogen receptor beta. Proc Natl Acad Sci U S A. 1998;95:15677–15682. doi: 10.1073/pnas.95.26.15677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Kriegsfeld LJ, Nelson RJ. Gonadal and photoperiodic influences on body mass regulation in adult male and female prairie voles. Am J Physiol. 1996;270:R1013–1018. doi: 10.1152/ajpregu.1996.270.5.R1013. [DOI] [PubMed] [Google Scholar]
  • 171.Krol E, Duncan JS, Redman P, Morgan PJ, Mercer JG, Speakman JR. Photoperiod regulates leptin sensitivity in field voles, Microtus agrestis. J Comp Physiol [B] 2006;176:153–163. doi: 10.1007/s00360-005-0037-8. [DOI] [PubMed] [Google Scholar]
  • 172.Landys MM, Goymann W, Schwabl I, Trapschuh M, Slagsvold T. Impact of season and social challenge on testosterone and corticosterone levels in a year-round territorial bird. Horm Behav. 2010;58:317–325. doi: 10.1016/j.yhbeh.2010.02.013. [DOI] [PubMed] [Google Scholar]
  • 173.Lavenex P, Steele MA, Jacobs LF. Sex differences, but no seasonal variations in the hippocampus of food-caching squirrels: a stereological study. J Comp Neurol. 2000;425:152–166. [PubMed] [Google Scholar]
  • 174.Lawlor DA, Davey Smith G, Mitchell R, Ebrahim S. Temperature at birth, coronary heart disease, and insulin resistance: cross sectional analyses of the British women's heart and health study. Heart. 2004;90:381–388. doi: 10.1136/hrt.2002.009548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Lee HJ, Macbeth AH, Pagani JH, Young WS., 3rd Oxytocin: the great facilitator of life. Prog Neurobiol. 2009;88:127–151. doi: 10.1016/j.pneurobio.2009.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Legan SJ, Karsch FJ, Foster DL. Endocrine Control of Seasonal Reproductive Function in Ewe - Marked Change in Response to Negative Feedback Action of Estradiol on Luteinizing-Hormone Secretion. Endocrinology. 1977;101:818–824. doi: 10.1210/endo-101-3-818. [DOI] [PubMed] [Google Scholar]
  • 177.Leitner C, Bartness TJ. Distributed forebrain sites mediate melatonin-induced short-day responses in Siberian hamsters. Endocrinology. 2010;151:3133–3140. doi: 10.1210/en.2010-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lendor C, Johnson A, Perzanowski M, Chew GL, Goldstein IF, Kelvin E, Perera F, Miller RL. Effects of winter birth season and prenatal cockroach and mouse allergen exposure on indoor allergen-specific cord blood mononuclear cell proliferation and cytokine production. Ann Allergy Asthma Immunol. 2008;101:193–199. doi: 10.1016/S1081-1206(10)60209-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Levine JE. Gonadotropin-Releasing Hormone (GnRH) In: Helen LH, Anthony WN, editors. Encyclopedia of Hormones. Academic Press; New York: 2003. pp. 157–165. [Google Scholar]
  • 180.Lewy AJ, Sack RL, Singer CM, White DM, Hoban TM. Winter depression and the phase-shift hypothesis for bright light's therapeutic effects: history, theory, and experimental evidence. J Biol Rhythms. 1988;3:121–134. doi: 10.1177/074873048800300203. [DOI] [PubMed] [Google Scholar]
  • 181.Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102:11070–11075. doi: 10.1073/pnas.0504978102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Ley RE, Hamady M, Lozupone C, Turnbaugh PJ, Ramey RR, Bircher JS, Schlegel ML, Tucker TA, Schrenzel MD, Knight R, Gordon JI. Evolution of mammals and their gut microbes. Science. 2008;320:1647–1651. doi: 10.1126/science.1155725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Li YF, Langholz B, Salam MT, Gilliland FD. Maternal and grandmaternal smoking patterns are associated with early childhood asthma. Chest. 2005;127:1232–1241. doi: 10.1378/chest.127.4.1232. [DOI] [PubMed] [Google Scholar]
  • 184.Lincoln G, Messager S, Andersson H, Hazlerigg D. Temporal expression of seven clock genes in the suprachiasmatic nucleus and the pars tuberalis of the sheep: evidence for an internal coincidence timer. Proc Natl Acad Sci U S A. 2002;99:13890–13895. doi: 10.1073/pnas.212517599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Liu J, Ballaney M, Al-alem U, Quan C, Jin X, Perera F, Chen LC, Miller RL. Combined inhaled diesel exhaust particles and allergen exposure alter methylation of T helper genes and IgE production in vivo. Toxicol Sci. 2008;102:76–81. doi: 10.1093/toxsci/kfm290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS, Smithies O. Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci U S A. 1993;90:11162–11166. doi: 10.1073/pnas.90.23.11162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Lucas RJ, Freedman MS, Munoz M, Garcia-Fernandez JM, Foster RG. Regulation of the mammalian pineal by non-rod, non-cone, ocular photoreceptors. Science. 1999;284:505–507. doi: 10.1126/science.284.5413.505. [DOI] [PubMed] [Google Scholar]
  • 188.Luedi PP, Hartemink AJ, Jirtle RL. Genome-wide prediction of imprinted murine genes. Genome Res. 2005;15:875–884. doi: 10.1101/gr.3303505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Lyte M. Microbial endocrinology and infectious disease in the 21st century. Trends Microbiol. 2004;12:14–20. doi: 10.1016/j.tim.2003.11.004. [DOI] [PubMed] [Google Scholar]
  • 190.Lyte M. The microbial organ in the gut as a driver of homeostasis and disease. Med Hypotheses. 2010;74:634–638. doi: 10.1016/j.mehy.2009.10.025. [DOI] [PubMed] [Google Scholar]
  • 191.Lyte M, Bailey MT. Neuroendocrine-bacterial interactions in a neurotoxin-induced model of trauma. J Surg Res. 1997;70:195–201. doi: 10.1006/jsre.1997.5130. [DOI] [PubMed] [Google Scholar]
  • 192.Madison DM, FitzGerald RW, McShea WJ. Dynamics of social nesting in overwintering meadow voles (Microtus pennsylvanicus): possible consequences for population cycling. Behav Ecol Sociobiol. 1984;15:9–14. [Google Scholar]
  • 193.Maestroni GJ, Conti A, Pierpaoli W. The pineal gland and the circadian, opiatergic, immunoregulatory role of melatonin. Ann N Y Acad Sci. 1987;496:67–77. doi: 10.1111/j.1749-6632.1987.tb35747.x. [DOI] [PubMed] [Google Scholar]
  • 194.Makino K, Nakamura H, Hide TI, Kuratsu JI. Risk of primary childhood brain tumors related to season of birth in Kumamoto Prefecture, Japan. Childs Nerv Syst. 2010 doi: 10.1007/s00381-010-1235-6. [DOI] [PubMed] [Google Scholar]
  • 195.Malpaux B, Migaud M, Tricoire H, Chemineau P. Biology of mammalian photoperiodism and the critical role of the pineal gland and melatonin. J Biol Rhythms. 2001;16:336–347. doi: 10.1177/074873001129002051. [DOI] [PubMed] [Google Scholar]
  • 196.Masana MI, Dubocovich ML. Melatonin receptor signaling: finding the path through the dark. Sci STKE 2001. 2001:pe39. doi: 10.1126/stke.2001.107.pe39. [DOI] [PubMed] [Google Scholar]
  • 197.Mason AO, Duffy S, Zhao S, Ubuka T, Bentley GE, Tsutsui K, Silver R, Kriegsfeld LJ. Photoperiod and reproductive condition are associated with changes in RFamide-related peptide (RFRP) expression in Syrian hamsters (Mesocricetus auratus) J Biol Rhythms. 2010;25:176–185. doi: 10.1177/0748730410368821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Mason AO, Greives TJ, Scotti MA, Levine J, Frommeyer S, Ketterson ED, Demas GE, Kriegsfeld LJ. Suppression of kisspeptin expression and gonadotropic axis sensitivity following exposure to inhibitory day lengths in female Siberian hamsters. Horm Behav. 2007;52:492–498. doi: 10.1016/j.yhbeh.2007.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Maywood ES, Hastings MH. Lesions of the iodomelatonin-binding sites of the mediobasal hypothalamus spare the lactotropic, but block the gonadotropic response of male Syrian hamsters to short photoperiod and to melatonin. Endocrinology. 1995;136:144–153. doi: 10.1210/endo.136.1.7828525. [DOI] [PubMed] [Google Scholar]
  • 200.McDowell TY, Amr S, Langenberg P, Royal W, Bever C, Culpepper WJ, Bradham DD. Time of birth, residential solar radiation and age at onset of multiple sclerosis. Neuroepidemiology. 2010;34:238–244. doi: 10.1159/000297749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Meitzen J, Thompson CK. Seasonal-like growth and regression of the avian song control system: neural and behavioral plasticity in adult male Gambel's white-crowned sparrows. Gen Comp Endocrinol. 2008;157:259–265. doi: 10.1016/j.ygcen.2008.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Mercer JG, Moar KM, Ross AW, Hoggard N, Morgan PJ. Photoperiod regulates arcuate nucleus POMC, AGRP, and leptin receptor mRNA in Siberian hamster hypothalamus. Am J Physiol. 2000;278:R271–281. doi: 10.1152/ajpregu.2000.278.1.R271. [DOI] [PubMed] [Google Scholar]
  • 203.Meredith JM, Turek FW, Levine JE. Effects of gonadotropin-releasing hormone pulse frequency modulation on the reproductive axis of photoinhibited male Siberian hamsters. Biol Reprod. 1998;59:813–819. doi: 10.1095/biolreprod59.4.813. [DOI] [PubMed] [Google Scholar]
  • 204.Messager S, Garabette ML, Hastings MH, Hazlerigg DG. Tissue-specific abolition of Per1 expression in the pars tuberalis by pinealectomy in the Syrian hamster. Neuroreport. 2001;12:579–582. doi: 10.1097/00001756-200103050-00029. [DOI] [PubMed] [Google Scholar]
  • 205.Messager S, Ross AW, Barrett P, Morgan PJ. Decoding photoperiodic time through Per1 and ICER gene amplitude. Proc Natl Acad Sci U S A. 1999;96:9938–9943. doi: 10.1073/pnas.96.17.9938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Mikulecky M, Cierna I. Seasonality of births and childhood inflammatory bowel disease. Wien Klin Wochenschr. 2005;117:554–557. doi: 10.1007/s00508-005-0391-2. [DOI] [PubMed] [Google Scholar]
  • 207.Molina-Hernandez M, Tellez-Alcantara P. Long photoperiod regimen may produce antidepressant actions in the male rat. Prog Neuropsychopharmacol Biol Psychiatry. 2000;24:105–116. doi: 10.1016/s0278-5846(99)00084-6. [DOI] [PubMed] [Google Scholar]
  • 208.Moore CB, Siopes TD. Enhancement of cellular and humoral immunity following embryonic exposure to melatonin in turkeys (Meleagris gallopavo) Gen Comp Endocrinol. 2005;143:178–183. doi: 10.1016/j.ygcen.2005.03.008. [DOI] [PubMed] [Google Scholar]
  • 209.Moore SE, Cole TJ, Poskitt EM, Sonko BJ, Whitehead RG, McGregor IA, Prentice AM. Season of birth predicts mortality in rural Gambia. Nature. 1997;388:434. doi: 10.1038/41245. [DOI] [PubMed] [Google Scholar]
  • 210.Morgan PJ, Barrett P, Howell HE, Helliwell R. Melatonin receptors: localization, molecular pharmacology and physiological significance. Neurochem Int. 1994;24:101–146. doi: 10.1016/0197-0186(94)90100-7. [DOI] [PubMed] [Google Scholar]
  • 211.Morgan PJ, Mercer JG. The regulation of body weight: lessons from the seasonal animal. Proc Nutr Soc. 2001;60:127–134. doi: 10.1079/pns200060. [DOI] [PubMed] [Google Scholar]
  • 212.Morgan PJ, Messager S, Webster C, Barrett P, Ross A. How does the melatonin receptor decode a photoperiodic signal in the pars tuberalis? Adv Exp Med Biol. 1999;460:165–174. doi: 10.1007/0-306-46814-x_18. [DOI] [PubMed] [Google Scholar]
  • 213.Murayama A, Sakura K, Nakama M, Yasuzawa-Tanaka K, Fujita E, Tateishi Y, Wang Y, Ushijima T, Baba T, Shibuya K, Shibuya A, Kawabe Y, Yanagisawa J. A specific CpG site demethylation in the human interleukin 2 gene promoter is an epigenetic memory. EMBO J. 2006;25:1081–1092. doi: 10.1038/sj.emboj.7601012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Nagy TR, Gower BA, Stetson MH. Response of collared lemmings to melatonin: I. Implants and photoperiod. J Pineal Res. 1994;17:177–184. doi: 10.1111/j.1600-079x.1994.tb00130.x. [DOI] [PubMed] [Google Scholar]
  • 215.Nakamura E, Kozaki K, Tsuda H, Suzuki E, Pimkhaokham A, Yamamoto G, Irie T, Tachikawa T, Amagasa T, Inazawa J, Imoto I. Frequent silencing of a putative tumor suppressor gene melatonin receptor 1 A (MTNR1A) in oral squamous-cell carcinoma. Cancer Sci. 2008;99:1390–1400. doi: 10.1111/j.1349-7006.2008.00838.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Nakao N, Ono H, Yamamura T, Anraku T, Takagi T, Higashi K, Yasuo S, Katou Y, Kageyama S, Uno Y, Kasukawa T, Iigo M, Sharp PJ, Iwasawa A, Suzuki Y, Sugano S, Niimi T, Mizutani M, Namikawa T, Ebihara S, Ueda HR, Yoshimura T. Thyrotrophin in the pars tuberalis triggers photoperiodic response. Nature. 2008;452:317–322. doi: 10.1038/nature06738. [DOI] [PubMed] [Google Scholar]
  • 217.Nelson RJ. Gonadal regression induced by caloric restriction is not mediated by the pineal gland in deer mice (Peromyscus maniculatus) J Pineal Res. 1987;4:339–345. doi: 10.1111/j.1600-079x.1987.tb00871.x. [DOI] [PubMed] [Google Scholar]
  • 218.Nelson RJ. Photoperiodic responsiveness in house mice. Physiol Behav. 1990;48:403–408. doi: 10.1016/0031-9384(90)90335-2. [DOI] [PubMed] [Google Scholar]
  • 219.Nelson RJ. Seasonal patterns of stress, immune function, and disease. Cambridge University Press; Cambridge, U.K; New York, NY: 2002. [Google Scholar]
  • 220.Nelson RJ. Seasonal immune function and sickness responses. Trends Immunol. 2004;25:187–192. doi: 10.1016/j.it.2004.02.001. [DOI] [PubMed] [Google Scholar]
  • 221.Nelson RJ. Biology of aggression. Oxford University Press; Oxford; New York: 2006. [Google Scholar]
  • 222.Nelson RJ, Demas GE. Seasonal changes in immune function. Q Rev Biol. 1996;71:511. doi: 10.1086/419555. [DOI] [PubMed] [Google Scholar]
  • 223.Nelson RJ, Demas GE, Huang PL, Fishman MC, Dawson VL, Dawson TM, Snyder SH. Behavioural abnormalities in male mice lacking neuronal nitric oxide synthase. Nature. 1995;378:383–386. doi: 10.1038/378383a0. [DOI] [PubMed] [Google Scholar]
  • 224.Nelson RJ, Demas GE, Klein SL, Kriegsfeld L. Seasonal patterns of stress, immune function and disease. Cambridge University Press; New York: 2002. [Google Scholar]
  • 225.Nelson RJ, Denlinger DL, Somers DE. Photoperiodism : The Biological Calendar. Oxford University Press; Oxford; New York: 2010. [Google Scholar]
  • 226.Nelson RJ, Kita M, Blom JM, Rhyne-Grey J. Photoperiod influences the critical caloric intake necessary to maintain reproduction among male deer mice (Peromyscus maniculatus) Biol Reprod. 1992;46:226–232. doi: 10.1095/biolreprod46.2.226. [DOI] [PubMed] [Google Scholar]
  • 227.Nelson RJ, Trainor BC. Neural mechanisms of aggression. Nat Rev Neurosci. 2007;8:536–546. doi: 10.1038/nrn2174. [DOI] [PubMed] [Google Scholar]
  • 228.Nelson RJ, Zucker I. Absence of Extra-Ocular Photoreception in Diurnal and Nocturnal Rodents Exposed to Direct Sunlight. Comp Biochem Physiol A Physiol. 1981;69:145–148. [Google Scholar]
  • 229.Nesse RM. Is depression an adaptation? Arch Gen Psychiatry. 2000;57:14–20. doi: 10.1001/archpsyc.57.1.14. [DOI] [PubMed] [Google Scholar]
  • 230.Nesse RM, Williams GC. Why we get sick: the new science of Darwinian medicine. 1st. Times Books; New York: 1994. [Google Scholar]
  • 231.Nosjean O, Ferro M, Coge F, Beauverger P, Henlin JM, Lefoulon F, Fauchere JL, Delagrange P, Canet E, Boutin JA. Identification of the melatonin-binding site MT3 as the quinone reductase 2. J Biol Chem. 2000;275:31311–31317. doi: 10.1074/jbc.M005141200. [DOI] [PubMed] [Google Scholar]
  • 232.Ono H, Hoshino Y, Yasuo S, Watanabe M, Nakane Y, Murai A, Ebihara S, Korf HW, Yoshimura T. Involvement of thyrotropin in photoperiodic signal transduction in mice. Proc Natl Acad Sci U S A. 2008;105:18238–18242. doi: 10.1073/pnas.0808952105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Pacheco AR, Sperandio V. Inter-kingdom signaling: chemical language between bacteria and host. Curr Opin Microbiol. 2009;12:192–198. doi: 10.1016/j.mib.2009.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Parker G, Walter S. Seasonal variation in depressive disorders and suicidal deaths in New South Wales. Br J Psychiatry. 1982;140:626–632. doi: 10.1192/bjp.140.6.626. [DOI] [PubMed] [Google Scholar]
  • 235.Parker KJ, Phillips KM, Kinney LF, Lee TM. Day length and sociosexual cohabitation alter central oxytocin receptor binding in female meadow voles (Microtus pennsylvanicus) Behav Neurosci. 2001;115:1349–1356. [PubMed] [Google Scholar]
  • 236.Paterson AT, Vickers C. Melatonin and the adrenal cortex: relationship to territorial aggression in mice. Physiol Behav. 1981;27:983–987. doi: 10.1016/0031-9384(81)90358-9. [DOI] [PubMed] [Google Scholar]
  • 237.Paul MJ, Pyter LM, Freeman DA, Galang J, Prendergast BJ. Photic and nonphotic seasonal cues differentially engage hypothalamic kisspeptin and RFamide-related peptide mRNA expression in Siberian hamsters. J Neuroendocrinol. 2009;21:1007–1014. doi: 10.1111/j.1365-2826.2009.01924.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Pelletier J, Bodin L, Hanocq E, Malpaux B, Teyssier J, Thimonier J, Chemineau P. Association between expression of reproductive seasonality and alleles of the gene for Mel(1a) receptor in the ewe. Biol Reprod. 2000;62:1096–1101. doi: 10.1095/biolreprod62.4.1096. [DOI] [PubMed] [Google Scholar]
  • 239.Pereira A, Rawson J, Jakubowska A, Clarke IJ. Estradiol-17beta-responsive A1 and A2 noradrenergic cells of the brain stem project to the bed nucleus of the stria terminalis in the ewe brain: a possible route for regulation of gonadotropin releasing hormone cells. Neuroscience. 2010;165:758–773. doi: 10.1016/j.neuroscience.2009.10.027. [DOI] [PubMed] [Google Scholar]
  • 240.Pfaff JJ, Bernert RA, Hollar DL, Witte TK, Merrill KA, Pettit JW, Almeida OP, Joiner TE., Jr Birth month and depressive and suicidal symptoms in an elderly Australian sample born in the Southern or Northern Hemisphere. Psychiatry Res. 2006;144:217–219. doi: 10.1016/j.psychres.2006.04.019. [DOI] [PubMed] [Google Scholar]
  • 241.Pompolo S, Pereira A, Scott CJ, Fujiyma F, Clarke IJ. Evidence for estrogenic regulation of gonadotropin-releasing hormone neurons by glutamatergic neurons in the ewe brain: An immunohistochemical study using an antibody against vesicular glutamate transporter-2. J Comp Neurol. 2003;465:136–144. doi: 10.1002/cne.10805. [DOI] [PubMed] [Google Scholar]
  • 242.Prendergast BJ. MT1 melatonin receptors mediate somatic, behavioral, and reproductive neuroendocrine responses to photoperiod and melatonin in Siberian hamsters (Phodopus sungorus) Endocrinology. 2010;151:714–721. doi: 10.1210/en.2009-0710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Prendergast BJ, Baillie SR, Dhabhar FS. Gonadal hormone-dependent and - independent regulation of immune function by photoperiod in Siberian hamsters. Am J Physiol. 2008;294:R384–392. doi: 10.1152/ajpregu.00551.2007. [DOI] [PubMed] [Google Scholar]
  • 244.Prendergast BJ, Bilbo SD, Nelson RJ. Photoperiod controls the induction, retention, and retrieval of antigen-specific immunological memory. Am J Physiol. 2004;286:R54–60. doi: 10.1152/ajpregu.00381.2003. [DOI] [PubMed] [Google Scholar]
  • 245.Prendergast BJ, Bilbo SD, Nelson RJ. Short day lengths enhance skin immune responses in gonadectomised Siberian hamsters. J Neuroendocrinol. 2005;17:18–21. doi: 10.1111/j.1365-2826.2005.01273.x. [DOI] [PubMed] [Google Scholar]
  • 246.Prendergast BJ, Gorman MR, Zucker I. Establishment and persistence of photoperiodic memory in hamsters. Proc Natl Acad Sci U S A. 2000;97:5586–5591. doi: 10.1073/pnas.100098597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Prendergast BJ, Hotchkiss AK, Bilbo SD, Kinsey SG, Nelson RJ. Photoperiodic adjustments in immune function protect Siberian hamsters from lethal endotoxemia. J Biol Rhythms. 2003;18:51. doi: 10.1177/0748730402239676. [DOI] [PubMed] [Google Scholar]
  • 248.Prendergast BJ, Kay LM. Affective and adrenocorticotrophic responses to photoperiod in Wistar rats. J Neuroendocrinol. 2008;20:261–267. doi: 10.1111/j.1365-2826.2007.01633.x. [DOI] [PubMed] [Google Scholar]
  • 249.Prendergast BJ, Nelson RJ. Affective responses to changes in day length in Siberian hamsters (Phodopus sungorus) Psychoneuroendocrinology. 2005;30:438–452. doi: 10.1016/j.psyneuen.2004.08.008. [DOI] [PubMed] [Google Scholar]
  • 250.Prendergast BJ, Pyter LM. Photoperiod history differentially impacts reproduction and immune function in adult Siberian hamsters. J Biol Rhythms. 2009;24:509–522. doi: 10.1177/0748730409349714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Prendergast BJ, Wynne-Edwards KE, Yellon SM, Nelson RJ. Photorefractoriness of immune function in male Siberian hamsters (Phodopus sungorus) J Neuroendocrinol. 2002;14:318–329. doi: 10.1046/j.1365-2826.2002.00781.x. [DOI] [PubMed] [Google Scholar]
  • 252.Prendergast BJ, Yellon SM, Tran LT, Nelson RJ. Photoperiod modulates the inhibitory effect of in vitro melatonin on lymphocyte proliferation in female Siberian hamsters. J Biol Rhythms. 2001;16:224–233. doi: 10.1177/074873040101600305. [DOI] [PubMed] [Google Scholar]
  • 253.Prendergast BJ, Zucker I, Nelson RJ. Seasonal rhythms of mammalian behavioral neuroendocrinology. In: Pfaff DW, editor. Hormones, brain, and behavior. Elsevier/Academic Press; Amsterdam; Boston: 2009. [Google Scholar]
  • 254.Prentice AM, Whitehead RG, Roberts SB, Paul AA. Long-term energy balance in child-bearing Gambian women. Am J Clin Nutr. 1981;34:2790–2799. doi: 10.1093/ajcn/34.12.2790. [DOI] [PubMed] [Google Scholar]
  • 255.Probst AV, Dunleavy E, Almouzni G. Epigenetic inheritance during the cell cycle. Nat Rev Mol Cell Biol. 2009;10:192–206. doi: 10.1038/nrm2640. [DOI] [PubMed] [Google Scholar]
  • 256.Ptashne M. On the use of the word ‘epigenetic’. Curr Biol. 2007;17:R233–236. doi: 10.1016/j.cub.2007.02.030. [DOI] [PubMed] [Google Scholar]
  • 257.Pyter LM, Adelson JD, Nelson RJ. Short days increase hypothalamic-pituitary-adrenal axis responsiveness. Endocrinology. 2007;148:3402–3409. doi: 10.1210/en.2006-1432. [DOI] [PubMed] [Google Scholar]
  • 258.Pyter LM, Nelson RJ. Enduring effects of photoperiod on affective behaviors in Siberian hamsters (Phodopus sungorus) Behav Neurosci. 2006;120:125–134. doi: 10.1037/0735-7044.120.1.125. [DOI] [PubMed] [Google Scholar]
  • 259.Pyter LM, Reader BF, Nelson RJ. Short photoperiods impair spatial learning and alter hippocampal dendritic morphology in adult male white-footed mice (Peromyscus leucopus) J Neurosci. 2005;25:4521–4526. doi: 10.1523/JNEUROSCI.0795-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Pyter LM, Weil ZM, Nelson RJ. Latitude affects photoperiod-induced changes in immune response in meadow voles (Microtus pennsylvanicus) Can J Zool. 2005;83:1271–1278. [Google Scholar]
  • 261.Reiter RJ, Tan DX, Fuentes-Broto L. Melatonin: a multitasking molecule. Prog Brain Res. 2010;181:127–151. doi: 10.1016/S0079-6123(08)81008-4. [DOI] [PubMed] [Google Scholar]
  • 262.Reppert SM, Godson C, Mahle CD, Weaver DR, Slaugenhaupt SA, Gusella JF. Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc Natl Acad Sci U S A. 1995;92:8734–8738. doi: 10.1073/pnas.92.19.8734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Reppert SM, Weaver DR, Ebisawa T, Mahle CD, Kolakowski LF., Jr Cloning of a melatonin-related receptor from human pituitary. FEBS Lett. 1996;386:219–224. doi: 10.1016/0014-5793(96)00437-1. [DOI] [PubMed] [Google Scholar]
  • 264.Revel FG, Saboureau M, Pevet P, Mikkelsen JD, Simonneaux V. Melatonin regulates type 2 deiodinase gene expression in the Syrian hamster. Endocrinology. 2006;147:4680–4687. doi: 10.1210/en.2006-0606. [DOI] [PubMed] [Google Scholar]
  • 265.Robinson JE, Karsch FJ. Refractoriness to inductive day lengths terminates the breeding season of the Suffolk ewe. Biol Reprod. 1984;31:656–663. doi: 10.1095/biolreprod31.4.656. [DOI] [PubMed] [Google Scholar]
  • 266.Romera EP, Mohamed F, Fogal T, Dominguez S, Piezzi R, Scardapane L. Effect of the photoperiod and administration of melatonin on the pars tuberalis of viscacha (Lagostomus maximus maximus): an ultrastructural study. Anat Rec. 2010;293:871–878. doi: 10.1002/ar.21083. [DOI] [PubMed] [Google Scholar]
  • 267.Ronchi E, Spencer RL, Krey LC, McEwen BS. Effects of photoperiod on brain corticosteroid receptors and the stress response in the golden hamster (Mesocricetus auratus) Brain Res. 1998;780:348–351. doi: 10.1016/s0006-8993(97)01303-6. [DOI] [PubMed] [Google Scholar]
  • 268.Rousseau K, Atcha Z, Cagampang FR, Le Rouzic P, Stirland JA, Ivanov TR, Ebling FJ, Klingenspor M, Loudon AS. Photoperiodic regulation of leptin resistance in the seasonally breeding Siberian hamster (Phodopus sungorus) Endocrinology. 2002;143:3083–3095. doi: 10.1210/endo.143.8.8967. [DOI] [PubMed] [Google Scholar]
  • 269.Rousseau K, Atcha Z, Loudon AS. Leptin and seasonal mammals. J Neuroendocrinol. 2003;15:409–414. doi: 10.1046/j.1365-2826.2003.01007.x. [DOI] [PubMed] [Google Scholar]
  • 270.Rubin RT, Dinan TG, Scott LV. The neuroendocrinology of affective disorders. Academic Press; New York: 2002. [Google Scholar]
  • 271.Sassi PL, Borghi CE, Bozinovic F. Spatial and seasonal plasticity in digestive morphology of cavies (Microcavia australis) inhabiting habitats with different plant qualities. J Mammal. 2007;88:165–172. [Google Scholar]
  • 272.Saxena N, Sinha MP. Pineal, Photoperiod and Gonadal Function in the Indian Palm Squirrel, Funambulus pennanti. Zoolog Sci. 2000;17:69–74. doi: 10.2108/zsj.17.69. [DOI] [PubMed] [Google Scholar]
  • 273.Schneider JE. Energy balance and reproduction. Physiol Behav. 2004;81:289–317. doi: 10.1016/j.physbeh.2004.02.007. [DOI] [PubMed] [Google Scholar]
  • 274.Schwabl H. Winter and Breeding Territorial Behavior and Levels of Reproductive Hormones of Migratory European Robins. Ornis Scandinavica. 1992;23:271–276. [Google Scholar]
  • 275.Schwartz WJ, de la Iglesia HO, Zlomanczuk P, Illnerova H. Encoding le quattro stagioni within the mammalian brain: photoperiodic orchestration through the suprachiasmatic nucleus. J Biol Rhythms. 2001;16:302–311. doi: 10.1177/074873001129002024. [DOI] [PubMed] [Google Scholar]
  • 276.Scotti MA, Place NJ, Demas GE. Short-day increases in aggression are independent of circulating gonadal steroids in female Siberian hamsters (Phodopus sungorus) Horm Behav. 2007;52:183–190. doi: 10.1016/j.yhbeh.2007.03.029. [DOI] [PubMed] [Google Scholar]
  • 277.Scotti MA, Schmidt KL, Newman AE, Bonu T, Soma KK, Demas GE. Aggressive encounters differentially affect serum dehydroepiandrosterone and testosterone concentrations in male Siberian hamsters (Phodopus sungorus) Horm Behav. 2009;56:376–381. doi: 10.1016/j.yhbeh.2009.07.004. [DOI] [PubMed] [Google Scholar]
  • 278.Segarra S, Mir R, Martinez C, Leon J. Genome-wide analyses of the transcriptomes of salicylic acid-deficient versus wild-type plants uncover Pathogen and Circadian Controlled 1 (PCC1) as a regulator of flowering time in Arabidopsis. Plant Cell Environ. 2010;33:11–22. doi: 10.1111/j.1365-3040.2009.02045.x. [DOI] [PubMed] [Google Scholar]
  • 279.Shaw D, Goldman BD. Influence of prenatal and postnatal photoperiods on postnatal testis development in the Siberian hamster (Phodopus sungorus) Biol Reprod. 1995;52:833–838. doi: 10.1095/biolreprod52.4.833. [DOI] [PubMed] [Google Scholar]
  • 280.Sherry DF, Hoshooley JS. Seasonal hippocampal plasticity in food-storing birds. Philos Trans R Soc Lond B Biol Sci. 2010;365:933–943. doi: 10.1098/rstb.2009.0220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Shimomura K, Lowrey PL, Vitaterna MH, Buhr ED, Kumar V, Hanna P, Omura C, Izumo M, Low SS, Barrett RK, LaRue SI, Green CB, Takahashi JS. Genetic suppression of the circadian Clock mutation by the melatonin biosynthesis pathway. Proc Natl Acad Sci U S A. 2010;107:8399–8403. doi: 10.1073/pnas.1004368107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Silva AL, Fry WH, Sweeney C, Trainor BC. Effects of photoperiod and experience on aggressive behavior in female California mice. Behav Brain Res. 2010;208:528–534. doi: 10.1016/j.bbr.2009.12.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Simonneaux V, Ansel L, Revel FG, Klosen P, Pevet P, Mikkelsen JD. Kisspeptin and the seasonal control of reproduction in hamsters. Peptides. 2009;30:146–153. doi: 10.1016/j.peptides.2008.06.006. [DOI] [PubMed] [Google Scholar]
  • 284.Simonneaux V, Ribelayga C. Generation of the melatonin endocrine message in mammals: a review of the complex regulation of melatonin synthesis by norepinephrine, peptides, and other pineal transmitters. Pharmacol Rev. 2003;55:325–395. doi: 10.1124/pr.55.2.2. [DOI] [PubMed] [Google Scholar]
  • 285.Singh K, Erdman RA, Swanson KM, Molenaar AJ, Maqbool NJ, Wheeler TT, Arias JA, Quinn-Walsh EC, Stelwagen K. Epigenetic regulation of milk production in dairy cows. J Mammary Gland Biol Neoplasia. 2010;15:101–112. doi: 10.1007/s10911-010-9164-2. [DOI] [PubMed] [Google Scholar]
  • 286.Sisk CL, Foster DL. The neural basis of puberty and adolescence. Nat Neurosci. 2004;7:1040–1047. doi: 10.1038/nn1326. [DOI] [PubMed] [Google Scholar]
  • 287.Skinner DC, Dufourny L. Oestrogen receptor beta-immunoreactive neurones in the ovine hypothalamus: distribution and colocalisation with gonadotropin-releasing hormone. J Neuroendocrinol. 2005;17:29–39. doi: 10.1111/j.1365-2826.2005.01271.x. [DOI] [PubMed] [Google Scholar]
  • 288.Smith JT, Clarke IJ. Kisspeptin expression in the brain: catalyst for the initiation of puberty. Rev Endocr Metab Disord. 2007;8:1–9. doi: 10.1007/s11154-007-9026-4. [DOI] [PubMed] [Google Scholar]
  • 289.Soma KK. Testosterone and aggression: Berthold, birds and beyond. J Neuroendocrinol. 2006;18:543–551. doi: 10.1111/j.1365-2826.2006.01440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Sotgiu S, Pugliatti M, Sotgiu MA, Fois ML, Arru G, Sanna A, Rosati G. Seasonal fluctuation of multiple sclerosis births in Sardinia. J Neurol. 2006;253:38–44. doi: 10.1007/s00415-005-0917-6. [DOI] [PubMed] [Google Scholar]
  • 291.Stearns SC. Life history evolution: successes, limitations, and prospects. Naturwissenschaften. 2000;87:476–486. doi: 10.1007/s001140050763. [DOI] [PubMed] [Google Scholar]
  • 292.Steinlechner S, Heldmaier G. Role of photoperiod and melatonin in seasonal acclimatization of the Djungarian hamster, Phodopus sungorus. Int J Biometeorol. 1982;26:329–337. doi: 10.1007/BF02219503. [DOI] [PubMed] [Google Scholar]
  • 293.Sumova A, Bendova Z, Sladek M, Kovacikova Z, Illnerova H. Seasonal molecular timekeeping within the rat circadian clock. Physiol Res. 2004;53 1:S167–176. [PubMed] [Google Scholar]
  • 294.Szyf M, Weaver IC, Champagne FA, Diorio J, Meaney MJ. Maternal programming of steroid receptor expression and phenotype through DNA methylation in the rat. Front Neuroendocrinol. 2005;26:139–162. doi: 10.1016/j.yfrne.2005.10.002. [DOI] [PubMed] [Google Scholar]
  • 295.Takahashi JS, Hong HK, Ko CH, McDearmon EL. The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat Rev Genet. 2008;9:764–775. doi: 10.1038/nrg2430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Tamarkin L, Hutchison JS, Goldman BD. Regulation of serum gonadotropins by photoperiod and testicular hormone in the Syrian hamster. Endocrinology. 1976;99:1528–1533. doi: 10.1210/endo-99-6-1528. [DOI] [PubMed] [Google Scholar]
  • 297.Tinbergen N. The Functions of Territory. Bird Study. 1957;4:14–27. [Google Scholar]
  • 298.Toloza EM, Lam M, Diamond J. Nutrient extraction by cold-exposed mice: a test of digestive safety margins. Am J Physiol. 1991;261:G608–620. doi: 10.1152/ajpgi.1991.261.4.G608. [DOI] [PubMed] [Google Scholar]
  • 299.Torrey EF, Miller J, Rawlings R, Yolken RH. Seasonality of births in schizophrenia and bipolar disorder: a review of the literature. Schizophr Res. 1997;28:1–38. doi: 10.1016/s0920-9964(97)00092-3. [DOI] [PubMed] [Google Scholar]
  • 300.Torrey EF, Miller J, Rawlings R, Yolken RH. Seasonal birth patterns of neurological disorders. Neuroepidemiology. 2000;19:177–185. doi: 10.1159/000026253. [DOI] [PubMed] [Google Scholar]
  • 301.Trainor BC, Lin S, Finy MS, Rowland MR, Nelson RJ. Photoperiod reverses the effects of estrogens on male aggression via genomic and nongenomic pathways. Proc Natl Acad Sci U S A. 2007;104:9840–9845. doi: 10.1073/pnas.0701819104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Trainor BC, Rowland MR, Nelson RJ. Photoperiod affects estrogen receptor alpha, estrogen receptor beta and aggressive behavior. Eur J Neurosci. 2007;26:207–218. doi: 10.1111/j.1460-9568.2007.05654.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Tramontin AD, Brenowitz EA. Seasonal plasticity in the adult brain. Trends Neurosci. 2000;23:251–258. doi: 10.1016/s0166-2236(00)01558-7. [DOI] [PubMed] [Google Scholar]
  • 304.Tsutsui K, Bentley GE, Bedecarrats G, Osugi T, Ubuka T, Kriegsfeld LJ. Gonadotropin-inhibitory hormone (GnIH) and its control of central and peripheral reproductive function. Front Neuroendocrinol. 2010;31:284–295. doi: 10.1016/j.yfrne.2010.03.001. [DOI] [PubMed] [Google Scholar]
  • 305.Tsutsui K, Saigoh E, Ukena K, Teranishi H, Fujisawa Y, Kikuchi M, Ishii S, Sharp PJ. A novel avian hypothalamic peptide inhibiting gonadotropin release. Biochem Biophys Res Commun. 2000;275:661–667. doi: 10.1006/bbrc.2000.3350. [DOI] [PubMed] [Google Scholar]
  • 306.Tups A, Ellis C, Moar KM, Logie TJ, Adam CL, Mercer JG, Klingenspor M. Photoperiodic regulation of leptin sensitivity in the Siberian hamster, Phodopus sungorus, is reflected in arcuate nucleus SOCS-3 (suppressor of cytokine signaling) gene expression. Endocrinology. 2004;145:1185–1193. doi: 10.1210/en.2003-1382. [DOI] [PubMed] [Google Scholar]
  • 307.Turek FW. The interaction of the photoperiod and testosterone in regulating serum gonadotropin levels in castrated male hamsters. Endocrinology. 1977;101:1210–1215. doi: 10.1210/endo-101-4-1210. [DOI] [PubMed] [Google Scholar]
  • 308.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
  • 309.Tuthill CR, Freeman DA, Butler MP, Chinn T, Park JH, Zucker I. Perinatal influences of melatonin on testicular development and photoperiodic memory in Siberian hamsters. J Neuroendocrinol. 2005;17:483–488. doi: 10.1111/j.1365-2826.2005.01329.x. [DOI] [PubMed] [Google Scholar]
  • 310.Ubuka T, Bentley GE, Ukena K, Wingfield JC, Tsutsui K. Melatonin induces the expression of gonadotropin-inhibitory hormone in the avian brain. Proc Natl Acad Sci U S A. 2005;102:3052–3057. doi: 10.1073/pnas.0403840102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Vaiserman AM, Khalangot MD, Carstensen B, Tronko MD, Kravchenko VI, Voitenko VP, Mechova LV, Koshel NM, Grigoriev PE. Seasonality of birth in adult type 2 diabetic patients in three Ukrainian regions. Diabetologia. 2009;52:2665–2667. doi: 10.1007/s00125-009-1519-0. [DOI] [PubMed] [Google Scholar]
  • 312.Valassi E, Scacchi M, Cavagnini F. Neuroendocrine control of food intake. Nutr Metab Cardiovasc Dis. 2008;18:158–168. doi: 10.1016/j.numecd.2007.06.004. [DOI] [PubMed] [Google Scholar]
  • 313.Van Ranst M, Joossens M, Joossens S, Van Steen K, Pierik M, Vermeire S, Rutgeerts P. Crohn's disease and month of birth. Inflamm Bowel Dis. 2005;11:597–599. doi: 10.1097/01.mib.0000163697.34592.d4. [DOI] [PubMed] [Google Scholar]
  • 314.Vassallo MF, Banerji A, Rudders SA, Clark S, Mullins RJ, Camargo CA., Jr Season of birth and food allergy in children. Ann Allergy Asthma Immunol. 2010;104:307–313. doi: 10.1016/j.anai.2010.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Veenema AH, Neumann ID. Central vasopressin and oxytocin release: regulation of complex social behaviours. Prog Brain Res. 2008;170:261–276. doi: 10.1016/S0079-6123(08)00422-6. [DOI] [PubMed] [Google Scholar]
  • 316.Vermeulen M, Palermo M, Giordano M. Neonatal pinealectomy impairs murine antibody-dependent cellular cytotoxicity. J Neuroimmunol. 1993;43:97–101. doi: 10.1016/0165-5728(93)90079-e. [DOI] [PubMed] [Google Scholar]
  • 317.von Gall C, Garabette ML, Kell CA, Frenzel S, Dehghani F, Schumm-Draeger PM, Weaver DR, Korf HW, Hastings MH, Stehle JH. Rhythmic gene expression in pituitary depends on heterologous sensitization by the neurohormone melatonin. Nat Neurosci. 2002;5:234–238. doi: 10.1038/nn806. [DOI] [PubMed] [Google Scholar]
  • 318.von Gall C, Stehle JH, Weaver DR. Mammalian melatonin receptors: molecular biology and signal transduction. Cell Tissue Res. 2002;309:151–162. doi: 10.1007/s00441-002-0581-4. [DOI] [PubMed] [Google Scholar]
  • 319.Wade GN, Bartness TJ. Effects of photoperiod and gonadectomy on food intake, body weight, and body composition in Siberian hamsters. Am J Physiol. 1984;246:R26–30. doi: 10.1152/ajpregu.1984.246.1.R26. [DOI] [PubMed] [Google Scholar]
  • 320.Waisman AS, Jacobs LF. Flexibility of cue use in the fox squirrel (Sciurus niger) Anim Cogn. 2008;11:625–636. doi: 10.1007/s10071-008-0152-5. [DOI] [PubMed] [Google Scholar]
  • 321.Walton JC, Chen Z, Weil ZM, Travers JB, Pyter LM, Nelson RJ. Photoperiod-mediated impairment of long term potentiation and learning and memory in male white-footed mice. Neuroscience. 2010 doi: 10.1016/j.neuroscience.2010.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Watanabe M, Yasuo S, Watanabe T, Yamamura T, Nakao N, Ebihara S, Yoshimura T. Photoperiodic regulation of type 2 deiodinase gene in Djungarian hamster: possible homologies between avian and mammalian photoperiodic regulation of reproduction. Endocrinology. 2004;145:1546–1549. doi: 10.1210/en.2003-1593. [DOI] [PubMed] [Google Scholar]
  • 323.Weaver DR, Keohan JT, Reppert SM. Definition of a prenatal sensitive period for maternal-fetal communication of day length. Am J Physiol. 1987;253:E701–704. doi: 10.1152/ajpendo.1987.253.6.E701. [DOI] [PubMed] [Google Scholar]
  • 324.Weaver DR, Liu C, Reppert SM. Nature's knockout: the Mel1b receptor is not necessary for reproductive and circadian responses to melatonin in Siberian hamsters. Mol Endocrinol. 1996;10:1478–1487. doi: 10.1210/mend.10.11.8923472. [DOI] [PubMed] [Google Scholar]
  • 325.Weaver IC, Cervoni N, Champagne FA, D'Alessio AC, Sharma S, Seckl JR, Dymov S, Szyf M, Meaney MJ. Epigenetic programming by maternal behavior. Nat Neurosci. 2004;7:847–854. doi: 10.1038/nn1276. [DOI] [PubMed] [Google Scholar]
  • 326.Wehr TA, Duncan WC, Jr, Sher L, Aeschbach D, Schwartz PJ, Turner EH, Postolache TT, Rosenthal NE. A circadian signal of change of season in patients with seasonal affective disorder. Arch Gen Psychiatry. 2001;58:1108–1114. doi: 10.1001/archpsyc.58.12.1108. [DOI] [PubMed] [Google Scholar]
  • 327.Weil ZM, Bowers SL, Nelson RJ. Photoperiod alters affective responses in collared lemmings. Behav Brain Res. 2007;179:305–309. doi: 10.1016/j.bbr.2007.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Weil ZM, Martin LB, 2nd, Nelson RJ. Photoperiod differentially affects immune function and reproduction in collared lemmings (Dicrostonyx groenlandicus) J Biol Rhythms. 2006;21:384–393. doi: 10.1177/0748730406292444. [DOI] [PubMed] [Google Scholar]
  • 329.Weil ZM, Norman GJ, DeVries AC, Berntson GG, Nelson RJ. Photoperiod alters autonomic regulation of the heart. Proc Natl Acad Sci U S A. 2009;106:4525–4530. doi: 10.1073/pnas.0810973106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Weil ZM, Pyter LM, Martin LB, 2nd, Nelson RJ. Perinatal photoperiod organizes adult immune responses in Siberian hamsters (Phodopus sungorus) Am J Physiol Regul Integr Comp Physiol. 2006;290:R1714–1719. doi: 10.1152/ajpregu.00869.2005. [DOI] [PubMed] [Google Scholar]
  • 331.Weil ZM, Workman JL, Nelson RJ. Housing condition alters immunological and reproductive responses to day length in Siberian hamsters (Phodopus sungorus) Horm Behav. 2007;52:261–266. doi: 10.1016/j.yhbeh.2007.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Wen JC, Hotchkiss AK, Demas GE, Nelson RJ. Photoperiod affects neuronal nitric oxide synthase and aggressive behaviour in male Siberian hamsters (Phodopus sungorus) J Neuroendocrinol. 2004;16:916–921. doi: 10.1111/j.1365-2826.2004.01248.x. [DOI] [PubMed] [Google Scholar]
  • 333.Wilkinson LS, Davies W, Isles AR. Genomic imprinting effects on brain development and function. Nat Rev Neurosci. 2007;8:832–843. doi: 10.1038/nrn2235. [DOI] [PubMed] [Google Scholar]
  • 334.Wingfield JC. A continuing saga: the role of testosterone in aggression. Horm Behav. 2005;48:253–255. doi: 10.1016/j.yhbeh.2005.05.009. discussion 256-258. [DOI] [PubMed] [Google Scholar]
  • 335.Wolff JO, Durr DS. Winter Nesting Behavior of Peromyscus leucopus and Peromyscus maniculatus. J Mammal. 1986;67:409–412. [Google Scholar]
  • 336.Woods SC, Seeley RJ. Adiposity signals and the control of energy homeostasis. Nutrition. 2000;16:894–902. doi: 10.1016/s0899-9007(00)00454-8. [DOI] [PubMed] [Google Scholar]
  • 337.Workman JL, Bowers SL, Nelson RJ. Enrichment and photoperiod interact to affect spatial learning and hippocampal dendritic morphology in white-footed mice (Peromyscus leucopus) Eur J Neurosci. 2009;29:161–170. doi: 10.1111/j.1460-9568.2008.06570.x. [DOI] [PubMed] [Google Scholar]
  • 338.Workman JL, Nelson RJ. Potential Animal Models of Seasonal Affective Disorder. Neurosci Biobehav Rev. 2010 doi: 10.1016/j.neubiorev.2010.08.005. [DOI] [PubMed] [Google Scholar]
  • 339.Workman JL, Weil ZM, Tuthill CR, Nelson RJ. Maternal pinealectomy increases depressive-like responses in Siberian hamster offspring. Behav Brain Res. 2008;189:387–391. doi: 10.1016/j.bbr.2008.01.016. [DOI] [PubMed] [Google Scholar]
  • 340.Xiong JJ, Karsch FJ, Lehman MN. Evidence for seasonal plasticity in the gonadotropin-releasing hormone (GnRH) system of the ewe: changes in synaptic inputs onto GnRH neurons. Endocrinology. 1997;138:1240–1250. doi: 10.1210/endo.138.3.5000. [DOI] [PubMed] [Google Scholar]
  • 341.Yaskin VA. Variation in brain morphology of the common shrew. 1994;18:155–161. Carnegie Museum of Natural History Special Publication. [Google Scholar]
  • 342.Yaskin VA. Seasonal adaptive modification of the hippocampus and dynamics of spatial behavior in bank voles (Clethrionomys glareolus, rodentia) Zool Zh. 2009;88:1365–1376. [Google Scholar]
  • 343.Yasuo S, Nakao N, Ohkura S, Iigo M, Hagiwara S, Goto A, Ando H, Yamamura T, Watanabe M, Watanabe T, Oda S, Maeda K, Lincoln GA, Okamura H, Ebihara S, Yoshimura T. Long-day suppressed expression of type 2 deiodinase gene in the mediobasal hypothalamus of the Saanen goat, a short-day breeder: implication for seasonal window of thyroid hormone action on reproductive neuroendocrine axis. Endocrinology. 2006;147:432–440. doi: 10.1210/en.2005-0507. [DOI] [PubMed] [Google Scholar]
  • 344.Yasuo S, Yoshimura T, Ebihara S, Korf HW. Temporal dynamics of type 2 deiodinase expression after melatonin injections in Syrian hamsters. Endocrinology. 2007;148:4385–4392. doi: 10.1210/en.2007-0497. [DOI] [PubMed] [Google Scholar]
  • 345.Yasuo S, Yoshimura T, Ebihara S, Korf HW. Photoperiodic control of TSH-beta expression in the mammalian pars tuberalis has different impacts on the induction and suppression of the hypothalamo-hypopysial gonadal axis. J Neuroendocrinol. 2010;22:43–50. doi: 10.1111/j.1365-2826.2009.01936.x. [DOI] [PubMed] [Google Scholar]
  • 346.Yellon SM, Teasley LA, Fagoaga OR, Nguyen HC, Truong HN, Nehlsen-Cannarella L. Role of photoperiod and the pineal gland in T cell-dependent humoral immune reactivity in the Siberian hamster. J Pineal Res. 1999;27:243–248. doi: 10.1111/j.1600-079x.1999.tb00622.x. [DOI] [PubMed] [Google Scholar]
  • 347.Yoshimura T, Yasuo S, Watanabe M, Iigo M, Yamamura T, Hirunagi K, Ebihara S. Light-induced hormone conversion of T4 to T3 regulates photoperiodic response of gonads in birds. Nature. 2003;426:178–181. doi: 10.1038/nature02117. [DOI] [PubMed] [Google Scholar]
  • 348.Zawilska JB, Skene DJ, Arendt J. Physiology and pharmacology of melatonin in relation to biological rhythms. Pharmacol Rep. 2009;61:383–410. doi: 10.1016/s1734-1140(09)70081-7. [DOI] [PubMed] [Google Scholar]
  • 349.Zhang JX, Zhang ZB, Wang ZW. Seasonal changes in and effects of familiarity on agonistic behaviors of rat-like hamsters (Cricetulus triton) Ecol Res. 2001;16:309–317. [Google Scholar]

RESOURCES