Elsevier

The Ocular Surface

Volume 20, April 2021, Pages 95-114
The Ocular Surface

High Impact Original Research
Light cycle phase advance as a model for jet lag reprograms the circadian rhythms of murine extraorbital lacrimal glands

https://doi.org/10.1016/j.jtos.2021.02.001Get rights and content

Abstract

Purpose

Jet lag causes a disruption in physiological rhythms in humans. This study aims to explore the extent to which jet lag affects the circadian rhythmicity in the lacrimal glands.

Methods

C57BL/6J mice were subjected to a 12-h light/12-h dark (LD) cycle and an 8-h advanced LD schedule as a model for jet lag. On day 5 after the LD advance, the extraorbital lacrimal glands (ELGs) were collected at 3-h intervals during a 24-h cycle. Total mRNA was extracted from normal and advanced LD-treated ELGs and assayed using high-throughput RNA sequencing. The rhythmic transcripts were identified, analyzed, and visualized by bioinformatics techniques. Finally, (i) animal behavior; (ii) the mass, cell size, and secretion response of ELGs; and (iii) circadian migration of immune cells to ELGs were also assayed.

Results

Jet lag treatment drastically altered the phase and composition of the rhythmic transcripts compared to that of normal ELGs. The key biological processes, signaling pathways, and protein–protein association networks were also dramatically altered in a spatiotemporal pattern. Furthermore, the circadian migration of neutrophils, T cells, B cells, and macrophages to the ELGs increased and shifted later by 6-h. Finally, the circadian rhythms of the ELGs with respect to mass, cell size, and secretion response were also impaired in jet lag-treated animals.

Conclusions

Jet lag impairs the circadian rhythm of the transcriptomic profile, structure, and secretion function of the lacrimal glands. This information provides novel insight into the negative effects of jet lag on ELGs.

Introduction

The tear film is the critical interface between the ocular surface and the external environment that maintains ocular surface homeostasis and corneal optical function [1]. Any factor that causes a change in the quality and quantity of the tear film can interfere with the imaging of external objects in the retina by decreasing contrast sensitivity and functional visual acuity. The tear film consists of three layers, namely, the lipid layer, aqueous layer, and mucin layer, from front to back. The aqueous layer is primarily derived from the secretion of the main lacrimal glands located in the orbital lacrimal fossa and the accessory lacrimal glands located in the palpebral and fornix conjunctivae. In addition to water and electrolytes, the aqueous layer also contains enzymes, such as peroxidase and lysozyme, lipids, lactoferrin, antimicrobial peptides [2], growth factors [3], lacritin [4], and cytokines [5]. These elements provide protection and support for the normal ocular surface through various mechanisms to avoid chemical, mechanical, and microbial damage from the outer environment [6].

The secretion of lacrimal glands is affected by a variety of internal and external factors [7]. Endogenous factors, such as menopause [8] and diabetes [9], reduce the secretion function of the lacrimal glands, which causes individuals to be prone to having dry eyes. Common external factors that affect secretion include cold, dryness, mechanical irritation, physical damage, toxic chemicals, and infections caused by various microbes [10]. However, whether and how alterations of the external light cycle disrupt the circadian clock system is not completely clear.

In mammals, virtually all aspects of physiological systems oscillate during the day under the control of the circadian clock system [11]. The synchronization of circadian oscillators is governed by the suprachiasmatic nucleus (SCN) in the hypothalamus, the central circadian pacemaker, after receiving light signal inputs from the retina [12]. Similarly, the metabolism and secretion of the lacrimal glands also have a robust rhythm during the day [[13], [14], [15]]. Human lacrimal glands secrete more tears during the daytime than at night [13]. Some tear film parameters, including pH [16], osmolarity [17,18], and tear volume [13,19], diurnally variate through the day. Circadian rhythm disturbances caused by shift work lead to instability of the tear film for night-shift workers and exacerbate dry eye symptoms [20]. The disturbance of normal circadian rhythms through sleep deprivation can also impair the structure and function of the lacrimal glands and, thus, induce dry eyes [21]. Moreover, we have systemically provided detailed information regarding the circadian characteristics of murine lacrimal glands with respect to cell size, secretion response, and transcriptomic profile [15,22]. We further demonstrated that metabolic stress induced by short-term high fructose intake [22] and streptozotocin-induced hyperglycemia [15] significantly reprograms the circadian rhythm of lacrimal glands at different levels.

Jet lag, which is caused by rapidly traversing numerous time zones, is a common desynchrony between an endogenous circadian clock and the external environment caused by shifts in the ambient light-dark cycle [[23], [24], [25]]. Acute jet lag can disrupt the normal circadian rhythm and lead to a series of psychological and physiological consequences, including sleep disturbances, daytime fatigue, decreased mental and physical performance, decreased alertness, headaches, and gastrointestinal disturbances [26,27]. However, to what extent jet lag affects the secretion response and molecular regulators of circadian rhythmicity in the lacrimal glands remains to be established.

Based on the discoveries described above, we hypothesized that circadian disruption by jet lag is sufficient to induce short-term lacrimal gland dysfunction, instability of tear film, and possible dry eye by fueling metabolic, immune, and neural dysfunction in the lacrimal glands. This study demonstrates that jet lag significantly influences alterations in the mass, cell size, secretion response, and circadian transcriptional profile of murine extraorbital lacrimal glands (ELGs) on a daily basis. These observations might contribute to our understanding of the molecular mechanism of the impairments in lacrimal gland physiology caused by jet lag as well as to the development of specific therapies for preventing jet lag-associated ocular surface diseases.

Section snippets

Overall study scheme and jet lag protocol

The flow chart of the experimental design and analysis for this study is presented in Fig. 1. In order to conduct this experiment, light cycle phase advance as a model for jet lag was established as previously described [26,28]. Briefly, C57BL/6J male mice were subjected to a 12-h light/12-h dark (LD) cycle for control group, and the LD cycle was advanced by 8-h for jet lag group (Fig. 1A). Locomotor activity and core body temperature were recorded as previously reported [26,28] (Fig. 1B). A

Jet lag alters general animal behavior

To determine the effects of jet lag on general murine phenotypes, the locomotor activity and core body temperature of the mice during light cycle phase advance as a model for jet lag were recorded by the Emitter-Telemetry System. As shown in Fig. 2A, the locomotor activity of the mice exhibited a diurnal rhythm, which increased during the dark phase, before the 8-h phase advance in the LD cycle. However, the locomotor activity of the mice gradually shifted after the changed light regimen, and

Discussion

To the best of our knowledge, this is the first report to confirm that light cycle phase advance as a model of jet lag globally disrupts the normal circadian rhythm of murine ELGs. This conclusion is based on the following observations: (1) the circadian patterns of murine ELGs in terms of weight, cell size, and secretion response after jet lag were significantly altered; (2) the composition of rhythmic genes and the phase of rhythmic gene expression were significantly altered in jet

Author contributions

Conceptualization, Z.L.; Methodology, S.H., D.Q., and X.P.; Investigation, X.J., D.L., X.P., D.Q., and S.H.; Analysis and visualization, S.H. and X.P.; Writing, Review and Editing, Z.L. and S.H.; Funding, Resources, and Supervision, Z.L., S.H., and X.J.

Declaration of competing interest

The authors declare no competing financial interest.

Acknowledgements

Research support was provided by the National Natural Science Foundation of China (grant numbers 81470603 and 81770962 to Z.L.), the Ministry of Science and Technology of the Peoples Republic of China (grant number 2018YFC0114500 to Z.L.), the Basic Science Project for Youth of Henan Eye Institute/Henan Eye Hospital (grant numbers 20JCQN003 to S.H. and 20JCQN002 to X.J.), the Doctoral Research and Development Foundation of Henan Provincial People's Hospital (grant number ZC20190146 to S.H.),

References (100)

  • J. Waterhouse et al.

    Jet lag: trends and coping strategies

    Lancet

    (2007)
  • A. Harkin et al.

    A study of VitalView for behavioural and physiological monitoring in laboratory rats

    Physiol Behav

    (2002)
  • M.D. Berquist et al.

    Phencyclidine-like in vivo effects of methoxetamine in mice and rats

    Neuropharmacology

    (2018)
  • D.R. Brown et al.

    A comparison of methods for organ-weight data adjustment in chicks

    Poultry Sci

    (1985)
  • Y. Zhang et al.

    RNA-sequencing based bone marrow cell transcriptome analysis reveals the potential mechanisms of E'jiao against blood-deficiency in mice

    Biomed Pharmacother

    (2019)
  • A. Chaix et al.

    Time-restricted feeding is a preventative and therapeutic intervention against diverse nutritional challenges

    Cell Metabol

    (2014)
  • M.E. Doyle et al.

    Autoimmune dacryoadenitis of NOD/LtJ mice and its subsequent effects on tear protein composition

    Am J Pathol

    (2007)
  • M. Baron et al.

    A single-cell transcriptomic map of the human and mouse pancreas reveals inter- and intra-cell population structure

    Cell Syst

    (2016)
  • C.A. Thaiss et al.

    Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis

    Cell

    (2014)
  • W. He et al.

    Circadian expression of migratory factors establishes lineage-specific signatures that guide the homing of leukocyte subsets to tissues

    Immunity

    (2018)
  • C. Scheiermann et al.

    Adrenergic nerves govern circadian leukocyte recruitment to tissues

    Immunity

    (2012)
  • T. Roenneberg et al.

    The circadian clock and human health

    Curr Biol

    (2016)
  • R. Wieczorek et al.

    The immunoarchitecture of the normal human lacrimal gland. Relevancy for understanding pathologic conditions

    Ophthalmology

    (1988)
  • J.E. Schechter et al.

    A lacrimal gland is a lacrimal gland, but rodent's and rabbit's are not human

    Ocul Surf

    (2010)
  • D.W. McGee et al.

    Lymphocyte migration into the lacrimal gland is random

    Cell Immunol

    (1984)
  • D. Druzd et al.

    Lymphocyte circadian clocks control lymph node trafficking and adaptive immune responses

    Immunity

    (2017)
  • K. Jin et al.

    Identification of lacrimal gland postganglionic innervation and its regulation of tear secretion

    Am J Pathol

    (2020)
  • R.M. Buijs et al.

    The circadian system and the balance of the autonomic nervous system

    Handb Clin Neurol

    (2013)
  • N. Gotlieb et al.

    Circadian control of neuroendocrine function: implications for health and disease

    Curr Opin Physiol

    (2018)
  • F. Baschieri et al.

    Circadian rhythms of cardiovascular autonomic function: physiology and clinical implications in neurodegenerative diseases

    Auton Neurosci

    (2019)
  • W.D. Todd et al.

    A time to fight: circadian control of aggression and associated autonomic support

    Auton Neurosci

    (2019)
  • C.L. Partch et al.

    Molecular architecture of the mammalian circadian clock

    Trends Cell Biol

    (2014)
  • B.G. Bunney et al.

    Mechanisms of rapid antidepressant effects of sleep deprivation therapy: clock genes and circadian rhythms

    Biol Psychiatr

    (2013)
  • P.H. Roseboom et al.

    Natural melatonin 'knockdown' in C57BL/6J mice: rare mechanism truncates serotonin N-acetyltransferase

    Brain Res Mol Brain Res

    (1998)
  • M.M. Macchi et al.

    Human pineal physiology and functional significance of melatonin

    Front Neuroendocrinol

    (2004)
  • P. Versura et al.

    Sex-steroid imbalance in females and dry eye

    Curr Eye Res

    (2015)
  • T.K. Yoo et al.

    Diabetes mellitus is associated with dry eye syndrome: a meta-analysis

    Int Ophthalmol

    (2019)
  • K.H. Cox et al.

    Circadian clock genes and the transcriptional architecture of the clock mechanism

    J Mol Endocrinol

    (2019)
  • M.H. Hastings et al.

    Generation of circadian rhythms in the suprachiasmatic nucleus

    Nat Rev Neurosci

    (2018)
  • A. Romano et al.

    The circadian rhythm of lacrimal secretion and its parameters, determined in a group of healthy individuals, and its potential diagnostic and therapeutic significance

    Adv Exp Med Biol

    (1994)
  • W.R. Webber et al.

    Fluorophotometric measurements of tear turnover rate in normal healthy persons: evidence for a circadian rhythm

    Eye

    (1987)
  • L.G. Carney et al.

    Human tear pH. Diurnal variations

    Arch Ophthalmol

    (1976)
  • W.J. Benjamin et al.

    Human tears: osmotic characteristics

    Invest Ophthalmol Vis Sci

    (1983)
  • J.E. Terry et al.

    Human tear osmotic pressure: diurnal variations and the closed eye

    Arch Ophthalmol

    (1978)
  • S. Srinivasan et al.

    Apparent time-dependent differences in inferior tear meniscus height in human subjects with mild dry eye symptoms

    Clin Exp Optom

    (2007)
  • S. Li et al.

    Sleep deprivation disrupts the lacrimal system and induces dry eye disease

    Exp Mol Med

    (2018)
  • D. Lu et al.

    Short-term high fructose intake reprograms the transcriptional clock rhythm of the murine extraorbital lacrimal gland

    Invest Ophthalmol Vis Sci

    (2019)
  • K.P. Wright et al.

    Intrinsic near-24-h pacemaker period determines limits of circadian entrainment to a weak synchronizer in humans

    Proc Natl Acad Sci USA

    (2001)
  • D.M. Berson et al.

    Phototransduction by retinal ganglion cells that set the circadian clock

    Science

    (2002)
  • S. Kiessling et al.

    Adrenal glucocorticoids have a key role in circadian resynchronization in a mouse model of jet lag

    J Clin Invest

    (2010)
  • Cited by (14)

    • Microbial Reconstitution Improves Aging-Driven Lacrimal Gland Circadian Dysfunction

      2021, American Journal of Pathology
      Citation Excerpt :

      The percentage of lipid staining (orange-red) was calculated relative to the whole lacrimal gland section area (n = 10). After a 4-week PBS or FMT treatment, mice (three per group) were euthanized by cervical dislocation every 3 hours during a circadian cycle (24 hours), and ELGs were collected as previously described.20,21,48 Total RNA was extracted from two lacrimal glands per animal using the Trizol RNA extraction protocol and purified using the RNeasy spin column kit (Qiagen, Hilden, Germany).

    View all citing articles on Scopus
    View full text