1
|
Korf HW, von Gall C. Mouse Models in Circadian Rhythm and Melatonin Research. J Pineal Res 2024; 76:e12986. [PMID: 38965880 DOI: 10.1111/jpi.12986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/06/2024]
Abstract
This contribution reviews the role of inbred and transgenic mouse strains for deciphering the mammalian melatoninergic and circadian system. It focusses on the pineal organ as melatonin factory and two major targets of the melatoninergic system, the suprachiasmatic nuclei (SCN) and the hypophysial pars tuberalis (PT). Mammalian pinealocytes sharing molecular characteristics with true pineal and retinal photoreceptors synthesize and secrete melatonin into the blood and cerebrospinal fluid night by night. Notably, neuron-like connections exist between the deep pinealocytes and the habenular/pretectal region suggesting direct pineal-brain communication. Control of melatonin biosynthesis in rodents involves transcriptional regulation including phosphorylation of CREB and upregulation of mPer1. In the SCN, melatonin acts upon MT1 and MT2 receptors. Melatonin is not necessary to maintain the rhythm of the SCN molecular clockwork, but it has distinct effects on the synchronization of the circadian rhythm by light, facilitates re-entrainment of the circadian system to phase advances in the level of the SCN molecular clockwork by acting upon MT2 receptors and plays a stabilizing role in the circadian system as evidenced from locomotor activity recordings. While the effects in the SCN are subtle, melatonin is essential for PT functions. Via the MT1 receptor it drives the PT-intrinsic molecular clockwork and the retrograde and anterograde output pathways controlling seasonal rhythmicity. Although inbred and transgenic mice do not show seasonal reproduction, the pathways from the PT are fully intact if the animals are melatonin proficient. Thus, only melatonin-proficient strains are suited to investigate the circadian and melatoninergic systems.
Collapse
Affiliation(s)
- Horst-Werner Korf
- Institute of Anatomy I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
2
|
Liu C, Yu H, Li Z, Chen S, Li X, Chen X, Chen B. The future of artificial hibernation medicine: protection of nerves and organs after spinal cord injury. Neural Regen Res 2024; 19:22-28. [PMID: 37488839 PMCID: PMC10479867 DOI: 10.4103/1673-5374.375305] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/05/2023] [Accepted: 04/17/2023] [Indexed: 07/26/2023] Open
Abstract
Spinal cord injury is a serious disease of the central nervous system involving irreversible nerve injury and various organ system injuries. At present, no effective clinical treatment exists. As one of the artificial hibernation techniques, mild hypothermia has preliminarily confirmed its clinical effect on spinal cord injury. However, its technical defects and barriers, along with serious clinical side effects, restrict its clinical application for spinal cord injury. Artificial hibernation is a future-oriented disruptive technology for human life support. It involves endogenous hibernation inducers and hibernation-related central neuromodulation that activate particular neurons, reduce the central constant temperature setting point, disrupt the normal constant body temperature, make the body "adapt" to the external cold environment, and reduce the physiological resistance to cold stimulation. Thus, studying the artificial hibernation mechanism may help develop new treatment strategies more suitable for clinical use than the cooling method of mild hypothermia technology. This review introduces artificial hibernation technologies, including mild hypothermia technology, hibernation inducers, and hibernation-related central neuromodulation technology. It summarizes the relevant research on hypothermia and hibernation for organ and nerve protection. These studies show that artificial hibernation technologies have therapeutic significance on nerve injury after spinal cord injury through inflammatory inhibition, immunosuppression, oxidative defense, and possible central protection. It also promotes the repair and protection of respiratory and digestive, cardiovascular, locomotor, urinary, and endocrine systems. This review provides new insights for the clinical treatment of nerve and multiple organ protection after spinal cord injury thanks to artificial hibernation. At present, artificial hibernation technology is not mature, and research faces various challenges. Nevertheless, the effort is worthwhile for the future development of medicine.
Collapse
Affiliation(s)
- Caiyun Liu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research Center of Experimental Acupucture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haixin Yu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research Center of Experimental Acupucture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhengchao Li
- Characteristic Medical Center of Chinese People’s Armed Police Force, Tianjin, China
| | - Shulian Chen
- Characteristic Medical Center of Chinese People’s Armed Police Force, Tianjin, China
| | - Xiaoyin Li
- Characteristic Medical Center of Chinese People’s Armed Police Force, Tianjin, China
| | - Xuyi Chen
- Characteristic Medical Center of Chinese People’s Armed Police Force, Tianjin, China
| | - Bo Chen
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research Center of Experimental Acupucture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Binhai New Area Hospital of TCM, Tianjin, China
- Fourth Teaching Hospital of Tianjin University of TCM, Tianjin, China
| |
Collapse
|
3
|
Roshan NJ, Garoussi MT, Akbarinejad V. Evaluation of the effect of melatonin implantation in rams and eCG dose in ewes synchronized by a CIDR-eCG protocol on reproductive performance of Lacaune sheep breed during non-breeding season. Anim Reprod Sci 2023; 259:107365. [PMID: 37980808 DOI: 10.1016/j.anireprosci.2023.107365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/02/2023] [Indexed: 11/21/2023]
Abstract
Sustainable production of milk is favorable in dairy sheep industry, which necessitates year-round reproduction of rams and ewes even during non-breeding season. Hence, protocols facilitating reproduction of rams and ewes during non-breeding season are of importance for this purpose. Therefore, the present study was conducted to investigate the effect of melatonin implantation in rams and administration of 400 IU eCG (E400) versus 300 IU eCG (E300) in ewes on reproductive performance of Lacaune sheep breed during non-breeding season. Rams were allocated to two groups including untreated (control, CON; n = 36) and melatonin-treated (MEL; n = 37). A subset of rams from CON (n = 7) and MEL (n = 7) groups were used for assessment of scrotal circumference, kinematic and functional characteristics of sperm, total antioxidant capacity (TAC) of semen and circulating testosterone on Days 0 (the beginning of study) and 60 (60 days after melatonin implantation). Further, the study had a 2 × 2 factorial design with four experimental groups including 1) ewes treated with E300 and introduced to CON rams (E300CON; n = 17 rams and 172 ewes), 2) ewes treated with E300 and introduced to MEL rams (E300MEL; n = 18 rams and 177 ewes), 3) ewes treated with E400 and introduced to CON rams (E400CON; n = 19 rams and 192 ewes), and ewes treated with E400 and introduced to MEL rams (E400MEL; n = 19 rams and 190 ewes). Melatonin implantation improved scrotal circumference, concentration, progressive motility, velocity, mitochondrial membrane potential and viability of sperm, TAC of semen, concentration of testosterone and fertility of rams (P < 0.05). Besides, E400 compared with E300 enhanced synchronization of estrus and fertility in ewes (P < 0.05). In conclusion, melatonin implantation promoted reproductive performance in Lacaune rams, and increase in dose of eCG improved reproductive performance in Lacaune ewes.
Collapse
Affiliation(s)
- Navid Jahan Roshan
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
4
|
Abulaiti A, Nawaz M, Naseer Z, Ahmed Z, Liu W, Abdelrahman M, Shaukat A, Sabek A, Pang X, Wang S. Administration of melatonin prior to modified synchronization protocol improves the productive and reproductive efficiency of Chinese crossbred buffaloes in low breeding season. Front Vet Sci 2023; 10:1118604. [PMID: 37261111 PMCID: PMC10228501 DOI: 10.3389/fvets.2023.1118604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/12/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Melatonin is a neurohormone involving various biological processes, including restoration of cyclicity in animals with seasonal breeding patterns. The use of melatonin in different forms has gained broader acceptance in different species, particularly in summer anestrous buffaloes. Objectives The objective of the current study was to evaluate the melatonin effect on the reproductive and productive performance of crossbred buffaloes during the low breeding season. Methods Sixty-five cyclic and reproductively sound crossbred buffaloes were randomly allocated to three groups: the G1 (n = 20) served as the control group and received no single melatonin, G2 received melatonin (n = 22; 18 mg/50 kg, body weight) once prior to synchronization and G3 group was administered multiple melatonin injections (n = 23; 6 mg/50 kg body weight) for three consecutive days before the start of the synchronization protocol. The reproductive performance, milk yield traits, and serum immunoglobulin M (IgM) and melatonin levels were evaluated in treated and untreated crossbred buffaloes. Results The results revealed that a single dose of melatonin administration has (p < 0.05) improved estrus response, ovulation occurrence and follicular growth in crossbred buffaloes compared to control groups. Higher pregnancy rates were observed in both melatonin-treated buffalo groups compared to the control. Following the administration of melatonin, serum IgM level increased in G2 and G3; however, an increment in melatonin level (p < 0.05) was detected in the G2 group only as compared to the control group subsequent day of melatonin administration. The milk compositions were not affected by melatonin administration except for milk urea nitrogen and somatic cell count (SCC). The melatonin administration (p < 0.05) decreased the somatic cell count in buffalo milk compared to untreated. Conclusion In conclusion, single or multiple doses of melatonin before initiating the synchronization protocol improved the ovulation, ovulatory follicle diameter and pregnancy rates in crossbred buffaloes during the low breeding season. Moreover, the administration of melatonin enhanced the IgM values along milk traits in terms of milk protein, MUN and somatic cell count in treated buffaloes.
Collapse
Affiliation(s)
- Adili Abulaiti
- College of Animal Science, Anhui Science and Technology University, Fengyang, AnHui, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang, AnHui, China
| | - Mudussar Nawaz
- Faculty of Veterinary and Animal Sciences, Pir Mehr Ali Shah Arid Agriculture University, Rawalpindi, Pakistan
| | - Zahid Naseer
- Faculty of Veterinary and Animal Sciences, Pir Mehr Ali Shah Arid Agriculture University, Rawalpindi, Pakistan
| | - Zulfiqar Ahmed
- Key Laboratory of Swine Genetics and Breeding, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan, China
| | - Wenju Liu
- College of Life and Health Science, Anhui Science and Technology University, Fengyang, AnHui, China
| | - Mohamed Abdelrahman
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan, China
| | - Aftab Shaukat
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan, China
| | - Ahmed Sabek
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Xunsheng Pang
- College of Animal Science, Anhui Science and Technology University, Fengyang, AnHui, China
| | - Shujuan Wang
- College of Animal Science, Anhui Science and Technology University, Fengyang, AnHui, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang, AnHui, China
| |
Collapse
|
5
|
Romanò N, Lafont C, Campos P, Guillou A, Fiordelisio T, Hodson DJ, Mollard P, Schaeffer M. Median eminence blood flow influences food intake by regulating ghrelin access to the metabolic brain. JCI Insight 2023; 8:165763. [PMID: 36574295 PMCID: PMC9977422 DOI: 10.1172/jci.insight.165763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Central integration of peripheral appetite-regulating signals ensures maintenance of energy homeostasis. Thus, plasticity of circulating molecule access to neuronal circuits involved in feeding behavior plays a key role in the adaptive response to metabolic changes. However, the mechanisms involved remain poorly understood despite their relevance for therapeutic development. Here, we investigated the role of median eminence mural cells, including smooth muscle cells and pericytes, in modulating gut hormone effects on orexigenic/anorexigenic circuits. We found that conditional activation of median eminence vascular cells impinged on local blood flow velocity and altered ghrelin-stimulated food intake by delaying ghrelin access to target neurons. Thus, activation of median eminence vascular cells modulates food intake in response to peripheral ghrelin by reducing local blood flow velocity and access to the metabolic brain.
Collapse
Affiliation(s)
| | - Chrystel Lafont
- Institute of Functional Genomics and,BioCampus Montpellier, University of Montpellier, CNRS, INSERM, F-34094 Montpellier, France
| | | | | | - Tatiana Fiordelisio
- Institute of Functional Genomics and,Laboratorio de Neuroendocrinología Comparada, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Science Facility, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David J. Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Patrice Mollard
- Institute of Functional Genomics and,BioCampus Montpellier, University of Montpellier, CNRS, INSERM, F-34094 Montpellier, France
| | - Marie Schaeffer
- Institute of Functional Genomics and,Centre de Biologie Structurale, CNRS UMR 5048, INSERM U1054, University of Montpellier, Montpellier, France
| |
Collapse
|
6
|
Liu B, Zhao R, Wu T, Ma Y, Gao Y, Wu Y, Hao B, Yin J, Li Y. Transcriptomes reveal microRNAs and mRNAs in different photoperiods influencing cashmere growth in goat. PLoS One 2023; 18:e0282772. [PMID: 36930617 PMCID: PMC10022811 DOI: 10.1371/journal.pone.0282772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
Cashmere goat has a typical characteristic in seasonal growth of cashmere. Studies have shown that one of the main factors affecting the cyclical growth of the cashmere is the photoperiod, however, its molecular mechanism remains unclear. Inner Mongolia Arbas cashmere goat was used to reveal the mRNA-microRNA regulatory mechanisms of cashmere growth in different photoperiod. Skin samples from cashmere goats under light control (short photoperiod) and normal conditions (long photoperiod) were collected. Sequencing was performed after RNA extraction. The differentially expressed miRNA and mRNA expression profiles were successfully constructed. We found 56 significantly differentially expressed known mRNAs (P<0.01) and 14 microRNAs (P<0.05). The association analysis of the microRNAs and mRNAs showed that two differentially expressed miRNAs might be targeted by six differentially expressed genes. Targeting relationships of these genes and miRNAs are revealed and verified. In all, the light control technology provides a new way to promote cashmere growth. Our results provide some references in the cashmere growth and development.
Collapse
Affiliation(s)
- Bin Liu
- Institute of Animal Husbandry, Academy of Agriculture and Stockbreeding Sciences, Hohhot, Inner Mongolia, China
| | - Ruoyang Zhao
- Institute of Animal Husbandry, Academy of Agriculture and Stockbreeding Sciences, Hohhot, Inner Mongolia, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory, Wenzhou, Wenzhou, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Tiecheng Wu
- Institute of Animal Husbandry, Academy of Agriculture and Stockbreeding Sciences, Hohhot, Inner Mongolia, China
| | - Yuejun Ma
- Institute of Animal Husbandry, Academy of Agriculture and Stockbreeding Sciences, Hohhot, Inner Mongolia, China
| | - Yulin Gao
- Institute of Animal Husbandry, Academy of Agriculture and Stockbreeding Sciences, Hohhot, Inner Mongolia, China
| | - Yahan Wu
- Institute of Animal Husbandry, Academy of Agriculture and Stockbreeding Sciences, Hohhot, Inner Mongolia, China
| | - Bayasihuliang Hao
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China
- Etuokeqianqi Arctic God Research Institute of Cashmere and Livestock, Erdos, China
| | - Jun Yin
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China
- * E-mail: (JY); (YL)
| | - Yurong Li
- Institute of Animal Husbandry, Academy of Agriculture and Stockbreeding Sciences, Hohhot, Inner Mongolia, China
- * E-mail: (JY); (YL)
| |
Collapse
|
7
|
Tortonese DJ. Hypophysial angiogenesis decodes annual time and underlies physiological adaptation to seasonal changes in the environment. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2022; 337:939-951. [PMID: 35844178 PMCID: PMC9796326 DOI: 10.1002/jez.2639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 01/01/2023]
Abstract
Adaptation to annual changes in the environment is controlled by hypophysial hormones. In temperate zones, photoperiod is the primary external cue that regulates annual biological cycles and is translated by the pattern of melatonin secretion acting primarily in the hypophysial pars tuberalis. Angiogenic mechanisms within this tissue contribute to decode the melatonin signal through alternative splicing of the vascular endothelial growth factor A (VEGF-A) gene in both the pars tuberalis and the capillary loops of the infundibulum. The resulting melatonin-evoked differential productions of VEGF-A isoforms will induce seasonal remodeling of the vascular connection between the hypothalamus and hypophysis, and act as paracrine messengers in the pars distalis to generate the required seasonal endocrine response. Specifically, the long melatonin signal in winter upregulates antiangiogenic VEGF-A isoforms, which will reduce the number of vascular loops and the density of VEGF receptors in endocrine and folliculo-stellate (FS) cells, inhibit prolactin secretion, and stimulate FSH. In contrast, the short melatonin signal in summer upregulates proangiogenic VEGF-A isoforms that will increase the number of vascular loops and the density of VEGF receptors in endocrine and FS cells, stimulate prolactin secretion, and suppress FSH. A similar system has been identified in long day seasonal breeders, revealing that this is a conserved mechanism of adaptation across species. Thus, an angiogenesis-based, intrahypophysial system for annual time measurement controls local microvascular plasticity and conveys the photoperiodic signal readout from the melatonin sensitive pars tuberalis to the endocrine cells of the pars distalis to regulate seasonal adaptation to the environment.
Collapse
Affiliation(s)
- Domingo J. Tortonese
- Laboratories for Integrative Neuroscience and Endocrinology, Faculty of Health SciencesUniversity of BristolBristolUK
| |
Collapse
|
8
|
Muacevic A, Adler JR, Eid FA, Alaswad HA, Ali WM, Aladraj FJ. Effects of Hormones and Endocrine Disorders on Hair Growth. Cureus 2022; 14:e32726. [PMID: 36578854 PMCID: PMC9788837 DOI: 10.7759/cureus.32726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Hormones have a close association with hair growth; thus, they have a big impact on the hair cycle and hair follicle structure. Many hormones control hair growth, cycle, and density. Hair abnormalities are frequent in therapeutic practice, and they can cause severe emotional discomfort depending on societal and ethnic standards. As a result, disorders that impact the endocrine system can induce a variety of physiological hair growth and cycling alterations. Hirsutism and patterned hair loss have a significant impact on human personality. These illnesses necessitate a comprehensive approach to diagnosis and treatment. The hormonal impact on hair growth and the association of different endocrine disorders with hair changes are briefly discussed here.
Collapse
|
9
|
Wei S, Kang X, Yang C, Wang F, Dai T, Guo X, Ma Z, Li C, Zhao H, Dan X. Analysis of reproduction-related transcriptomes on pineal-hypothalamic-pituitary-ovarian tissues during estrus and anestrus in Tan sheep. Front Vet Sci 2022; 9:1068882. [PMID: 36504859 PMCID: PMC9729709 DOI: 10.3389/fvets.2022.1068882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Seasonal estrus is an important factor limiting the fertility of some animals such as sheep. Promoting estrus in the anestrus season is one of the major ways in improving the fecundity of seasonally breeding animals. The pineal-hypothalamus-pituitary-ovary (PHPO) axis plays a decisive role in regulating animal reproduction. However, the molecular mechanisms by which the PHPO axis regulates seasonal reproduction in animals are not well understood, especially in Tan sheep. To this end, we collected pineal, hypothalamus, pituitary and ovary tissues from Tan sheep during estrus and anestrus for RNA-Sequencing, and performed bioinformatics analysis on the entire regulatory axis of the pineal-hypothalamic-pituitary-ovary (PHPO). The results showed that 940, 1,638, 750, and 971 DEGs (differentially expressed genes, DEGs) were identified in pineal, hypothalamus, pituitary and ovary, respectively. GO analysis showed that DEGs from PHPO axis-related tissues were mainly enriched in "biological processes" such as transmembrane transport, peptide and amide biosynthesis and DNA synthesis. Meanwhile, KEGG enrichment analysis showed that the bile acid secretion pathway and the neuroactive ligand-receptor interaction pathway were significantly enriched. Additionally, four potential candidate genes related to seasonal reproduction (VEGFA, CDC20, ASPM, and PLCG2) were identified by gene expression profiling and protein-protein interaction (PPI) analysis. These findings will contribute to be better understanding of seasonal reproduction regulation in Tan sheep and will serve as a useful reference for molecular breeding of high fertility Tan sheep.
Collapse
|
10
|
Le Tissier PR, Murray JF, Mollard P. A New Perspective on Regulation of Pituitary Plasticity: The Network of SOX2-Positive Cells May Coordinate Responses to Challenge. Endocrinology 2022; 163:6609891. [PMID: 35713880 PMCID: PMC9273012 DOI: 10.1210/endocr/bqac089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Indexed: 11/19/2022]
Abstract
Plasticity of function is required for each of the anterior pituitary endocrine axes to support alterations in the demand for hormone with physiological status and in response to environmental challenge. This plasticity is mediated at the pituitary level by a change in functional cell mass resulting from a combination of alteration in the proportion of responding cells, the amount of hormone secreted from each cell, and the total number of cells within an endocrine cell population. The functional cell mass also depends on its organization into structural and functional networks. The mechanisms underlying alteration in gland output depend on the strength of the stimulus and are axis dependent but in all cases rely on sensing of output of the functional cell mass and its regulation. Here, we present evidence that the size of pituitary cell populations is constrained and suggest this is mediated by a form of quorum sensing. We propose that pituitary cell quorum sensing is mediated by interactions between the networks of endocrine cells and hormone-negative SOX2-positive (SOX2+ve) cells and speculate that the latter act as both a sentinel and actuator of cell number. Evidence for a role of the network of SOX2+ve cells in directly regulating secretion from multiple endocrine cell networks suggests that it also regulates other aspects of the endocrine cell functional mass. A decision-making role of SOX2+ve cells would allow precise coordination of pituitary axes, essential for their appropriate response to physiological status and challenge, as well as prioritization of axis modification.
Collapse
Affiliation(s)
- Paul R Le Tissier
- Correspondence: Paul R. Le Tissier, PhD, Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Bldg, 15 George Square, Edinburgh EH8 9XD, UK.
| | - Joanne F Murray
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Patrice Mollard
- Correspondence: Patrice Mollard, PhD, Institute of Functional Genomics, University of Montpellier, 141 rue de la Cardonille, F-34093, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
11
|
Aubry LM, Williams CT. Vertebrate Phenological Plasticity: from Molecular Mechanisms to Ecological and Evolutionary Implications. Integr Comp Biol 2022; 62:958-971. [PMID: 35867980 DOI: 10.1093/icb/icac121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/12/2022] Open
Abstract
Seasonal variation in the availability of essential resources is one of the most important drivers of natural selection on the phasing and duration of annually recurring life-cycle events. Shifts in seasonal timing are among the most commonly reported responses to climate change and the capacity of organisms to adjust their timing, either through phenotypic plasticity or evolution, is a critical component of resilience. Despite growing interest in documenting and forecasting the impacts of climate change on phenology, our ability to predict how individuals, populations, and species might alter their seasonal timing in response to their changing environments is constrained by limited knowledge regarding the cues animals use to adjust timing, the endogenous genetic and molecular mechanisms that transduce cues into neural and endocrine signals, and the inherent capacity of animals to alter their timing and phasing within annual cycles. Further, the fitness consequences of phenological responses are often due to biotic interactions within and across trophic levels, rather than being simple outcomes of responses to changes in the abiotic environment. Here, we review the current state of knowledge regarding the mechanisms that control seasonal timing in vertebrates, as well as the ecological and evolutionary consequences of individual, population, and species-level variation in phenological responsiveness. Understanding the causes and consequences of climate-driven phenological shifts requires combining ecological, evolutionary, and mechanistic approaches at individual, populational, and community scales. Thus, to make progress in forecasting phenological responses and demographic consequences, we need to further develop interdisciplinary networks focused on climate change science.
Collapse
Affiliation(s)
- Lise M Aubry
- Department of Fish, Wildlife, and Conservation Biology, Colorado State University, 1474 Campus Delivery, Fort Collins, CO, 80523, USA
| | - Cory T Williams
- Department of Biology, Colorado State University, 1878 Campus Delivery Fort Collins, CO 80523, USA
| |
Collapse
|
12
|
Matsuoka RL, Buck LD, Vajrala KP, Quick RE, Card OA. Historical and current perspectives on blood endothelial cell heterogeneity in the brain. Cell Mol Life Sci 2022; 79:372. [PMID: 35726097 PMCID: PMC9209386 DOI: 10.1007/s00018-022-04403-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Dynamic brain activity requires timely communications between the brain parenchyma and circulating blood. Brain-blood communication is facilitated by intricate networks of brain vasculature, which display striking heterogeneity in structure and function. This vascular cell heterogeneity in the brain is fundamental to mediating diverse brain functions and has long been recognized. However, the molecular basis of this biological phenomenon has only recently begun to be elucidated. Over the past century, various animal species and in vitro systems have contributed to the accumulation of our fundamental and phylogenetic knowledge about brain vasculature, collectively advancing this research field. Historically, dye tracer and microscopic observations have provided valuable insights into the anatomical and functional properties of vasculature across the brain, and these techniques remain an important approach. Additionally, recent advances in molecular genetics and omics technologies have revealed significant molecular heterogeneity within brain endothelial and perivascular cell types. The combination of these conventional and modern approaches has enabled us to identify phenotypic differences between healthy and abnormal conditions at the single-cell level. Accordingly, our understanding of brain vascular cell states during physiological, pathological, and aging processes has rapidly expanded. In this review, we summarize major historical advances and current knowledge on blood endothelial cell heterogeneity in the brain, and discuss important unsolved questions in the field.
Collapse
Affiliation(s)
- Ryota L Matsuoka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Luke D Buck
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Keerti P Vajrala
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.,Kansas City University College of Osteopathic Medicine, Kansas City, MO 64106, USA
| | - Rachael E Quick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Olivia A Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| |
Collapse
|
13
|
Chevillard PM, Batailler M, Piégu B, Estienne A, Blache MC, Dubois JP, Pillon D, Vaudin P, Dupont J, Just N, Migaud M. Seasonal vascular plasticity in the mediobasal hypothalamus of the adult ewe. Histochem Cell Biol 2022; 157:581-593. [PMID: 35118552 DOI: 10.1007/s00418-022-02079-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
Abstract
Sheep, like most seasonal mammals, exhibit a cyclic adaptive reproductive physiology that allows ewes to give birth to their progeny during the spring when environmental conditions are favorable to their survival. This process relies on the detection of day length (or photoperiod) and is associated with profound changes in cellular plasticity and gene expression in the hypothalamic-pituitary-gonadal axis, mechanisms that are suggested to participate in the seasonal adaptation of neuroendocrine circuits. Recently, pituitary vascular growth has been proposed as a seasonally regulated process in which the vascular endothelial growth factor A (VEGFA), a well-known angiogenic cytokine, is suspected to play a crucial role. However, whether this mechanism is restricted to the pituitary gland or also occurs in the mediobasal hypothalamus (MBH), a crucial contributor to the control of the reproductive function, remains unexplored. Using newly developed image analysis tools, we showed that the arcuate nucleus (ARH) of the MBH exhibits an enhanced vascular density during the long photoperiod or non-breeding season, associated with higher expression of VEGFA. In the median eminence (ME), a structure connecting the MBH to the pituitary gland, higher VEGFA, kinase insert domain receptor (KDR/VEGFR2) and plasmalemma vesicle-associated protein (PLVAP) gene expressions were detected during the long photoperiod. We also found that VEGFA and its receptor, VEGFR2, are expressed by neurons and tanycytes in both the ARH and ME. Altogether, these data show variations in the MBH vasculature according to seasons potentially through a VEGFA-dependent pathway, paving the way for future studies aiming to decipher the role of these changes in the hypothalamic control of seasonal reproduction.
Collapse
Affiliation(s)
- Pierre-Marie Chevillard
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Martine Batailler
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Benoît Piégu
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Anthony Estienne
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Marie-Claire Blache
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Jean-Philippe Dubois
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Delphine Pillon
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Pascal Vaudin
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Joëlle Dupont
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Nathalie Just
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France
| | - Martine Migaud
- Physiologie de la Reproduction et des Comportements PRC Centre INRAE Val de Loire, CNRS, IFCE, INRAE, Université de Tours, 37380, Nouzilly, France.
| |
Collapse
|
14
|
Le Tissier PR, Mollard P. Renewing an old interest: Pituitary folliculostellate cells. J Neuroendocrinol 2021; 33:e13053. [PMID: 34734454 PMCID: PMC11475256 DOI: 10.1111/jne.13053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022]
Abstract
Anterior pituitary folliculostellate (FS) cells, first described almost 50 years ago, have a wide range of functions with respect to supporting and coordinating endocrine cell function, in particular through paracrine and gap junction-mediated signalling. Our previous studies identified the morphological organisation of FS cells, which mediates coordinated calcium activity throughout the homotypic FS network and allows signalling across the whole pituitary gland. It is also clear that FS cells can modify endocrine output and feedback on pituitary axes over a range of timescales. Recently, several studies have defined FS cells as a source of anterior pituitary endocrine cell renewal, which has resulted in a renaming of FS cells as "Sox2+ve stem cells". Here, we highlight the broader potential of the FS cell population in fine-tuning and coordinating pituitary axes function. In addition, we identify a need for: the definition of the possible subtypes of FS cell and their relationship with the stem cell population; the potential role of FS cells in pulsatile hormone secretion and coordination of heterotypic cell networks; and the roles that FS cells may play in both early-life programming of pituitary axes and in memory, or anticipation, of demand. Further studies of FS cells may demonstrate the fundamental importance of this cell type and its potential as a therapeutic target to correct pituitary gland dysfunction, one of which is stem cell therapy. Clearly, a thorough understanding of all of these interactions and relationships of FS and endocrine cells is required whatever therapeutic use is suggested by their various roles.
Collapse
Affiliation(s)
| | - Patrice Mollard
- Institute of Functional GenomicsCNRS, INSERMUniversity of MontpellierMontpellierFrance
| |
Collapse
|
15
|
El-Shalofy A, Hedia M, Kastelic J. Melatonin improves testicular haemodynamics, echotexture and testosterone production in Ossimi rams during the breeding season. Reprod Domest Anim 2021; 56:1456-1463. [PMID: 34459033 DOI: 10.1111/rda.14010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022]
Abstract
The objective was to determine effects of a single parenteral dose of melatonin on testicular blood flow indices, testicular echogenicity and plasma testosterone concentrations in rams during the physiological breeding season. We hypothesized that melatonin enhances testicular blood flow, echogenicity and plasma testosterone concentrations during the breeding season in rams. During the breeding season, 12 sexually mature Ossimi rams were randomly allocated to either a melatonin group (n = 8) that received 18 mg of melatonin in 1 ml of corn oil (injected SC) or a control group (n = 4) that received 1 ml corn oil only. Blood collection and ultrasonographic assessment of the testes and supratesticular arteries were conducted immediately before treatment (W0) and once weekly for 6 weeks after melatonin injection (W1-W6). Mean plasma testosterone concentrations were greater (p < .05; at least 1 ng/ml) in the melatonin-treated group compared to the control group from W4 to W6 after treatment. A decrease (p < .05) in both resistive index (RI) and pulsatility index (PI) began 1 week after melatonin injection (W1) and persisted until the end of the experiment, with mean RI and PI values in the melatonin group lower (p < .05) than those in the control group on W3 and W4. Furthermore, plasma testosterone concentrations in melatonin-treated rams were inversely correlated to both RI and PI (r = -.7 and -.6, respectively, p < .01). Testicular echogenicity decreased (p < .05) 1 week after melatonin injection (W1) and remained lower (p < .05) in the melatonin-treated group compared to the control group until the end of the study (W6). In conclusion, melatonin administration significantly altered testicular blood flow and echogenicity and increased plasma testosterone concentrations in Ossimi rams during the breeding season.
Collapse
Affiliation(s)
- Amr El-Shalofy
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mohamed Hedia
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - John Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
16
|
Star E, Stevens M, Gooding C, Smith CWJ, Li L, Ayine ML, Harper SJ, Bates DO, Oltean S. A drug-repositioning screen using splicing-sensitive fluorescent reporters identifies novel modulators of VEGF-A splicing with anti-angiogenic properties. Oncogenesis 2021; 10:36. [PMID: 33941763 PMCID: PMC8093282 DOI: 10.1038/s41389-021-00323-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 03/25/2021] [Accepted: 04/09/2021] [Indexed: 12/30/2022] Open
Abstract
Alternative splicing of the vascular endothelial growth factor A (VEGF-A) terminal exon generates two protein families with differing functions. Pro-angiogenic VEGF-Axxxa isoforms are produced via selection of the proximal 3' splice site of the terminal exon. Use of an alternative distal splice site generates the anti-angiogenic VEGF-Axxxb proteins. A bichromatic splicing-sensitive reporter was designed to mimic VEGF-A alternative splicing and was used as a molecular tool to further investigate this alternative splicing event. Part of VEGF-A's terminal exon and preceding intron were inserted into a minigene construct followed by the coding sequences for two fluorescent proteins. A different fluorescent protein is expressed depending on which 3' splice site of the exon is used during splicing (dsRED denotes VEGF-Axxxa and EGFP denotes VEGF-Axxxb). The fluorescent output can be used to follow splicing decisions in vitro and in vivo. Following successful reporter validation in different cell lines and altering splicing using known modulators, a screen was performed using the LOPAC library of small molecules. Alterations to reporter splicing were measured using a fluorescent plate reader to detect dsRED and EGFP expression. Compounds of interest were further validated using flow cytometry and assessed for effects on endogenous VEGF-A alternative splicing at the mRNA and protein level. Ex vivo and in vitro angiogenesis assays were used to demonstrate the anti-angiogenic effect of the compounds. Furthermore, anti-angiogenic activity was investigated in a Matrigel in vivo model. To conclude, we have identified a set of compounds that have anti-angiogenic activity through modulation of VEGF-A terminal exon splicing.
Collapse
Affiliation(s)
- Eleanor Star
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Megan Stevens
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Clare Gooding
- grid.5335.00000000121885934Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW UK
| | - Christopher W. J. Smith
- grid.5335.00000000121885934Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW UK
| | - Ling Li
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Monica Lamici Ayine
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - Steve J. Harper
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | - David O. Bates
- grid.415598.40000 0004 0641 4263Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen’s Medical Centre, West Block, D floor, Nottingham, NG7 2UH UK
| | - Sebastian Oltean
- grid.8391.30000 0004 1936 8024Institute of Biomedical & Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| |
Collapse
|
17
|
Tanycytes in the infundibular nucleus and median eminence and their role in the blood-brain barrier. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:253-273. [PMID: 34225934 DOI: 10.1016/b978-0-12-820107-7.00016-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier is generally attributed to endothelial cells. However, in circumventricular organs, such as the median eminence, tanycytes take over the barrier function. These ependymoglial cells form the wall of the third ventricle and send long extensions into the parenchyma to contact blood vessels and hypothalamic neurons. The shape and location of tanycytes put them in an ideal position to connect the periphery with central nervous compartments. In line with this, tanycytes control the transport of hormones and key metabolites in and out of the hypothalamus. They function as sensors of peripheral homeostasis for central regulatory networks. This chapter discusses current evidence that tanycytes play a key role in regulating glucose balance, food intake, endocrine axes, seasonal changes, reproductive function, and aging. The understanding of how tanycytes perform these diverse tasks is only just beginning to emerge and will probably lead to a more differentiated view of how the brain and the periphery interact.
Collapse
|
18
|
Vázquez-Membrillo M, Siqueiros-Márquez L, Núñez FF, Díaz-Lezama N, Adán-Castro E, Ramírez-Hernández G, Adán N, Macotela Y, Martínez de la Escalera G, Clapp C. Prolactin stimulates the vascularisation of the retina in newborn mice under hyperoxia conditions. J Neuroendocrinol 2020; 32:e12858. [PMID: 32449569 DOI: 10.1111/jne.12858] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/17/2020] [Accepted: 04/21/2020] [Indexed: 12/23/2022]
Abstract
The hormone prolactin (PRL) is emerging as an important regulator of ocular blood vessels. PRL is pro-angiogenic and acquires anti-angiogenic properties after undergoing proteolytic cleavage to the PRL fragment, vasoinhibin. The vascularisation of the rodent retina develops after birth when it rapidly expands until completion at the end of the first postnatal week. Exposure of newborn mice to high oxygen levels lowers the rate of blood vessel growth. In the present study, we investigated whether PRL treatment modifies the vascularisation of the retina in newborn mice exposed to high oxygen or to normoxia and whether the retinal conversion of PRL to vasoinhibin may be altered in the neonate. Newborn mice and their nursing mothers were subjected to 75% oxygen or to normoxia from postnatal day (P) 6 to P8 (group 1) or from P2 to P5 (group 2). PRL (2 µg g-1 , i.p., twice a day) or vehicle was injected from P5 to P8 in group 1 and from P1 to P5 in group 2. PRL treatment reduced the retinal inhibition of blood vessel growth and the increase in vascular regression induced by hyperoxia as revealed by immunofluorescence staining of blood vessels and the expression of angiogenesis and apoptosis markers. The pro-angiogenic effect may involve a reduced conversion of PRL to vasoinhibin. Incubation of PRL with retinal extracts showed reduced activity of the PRL-cleaving protease, cathepsin D, in the neonate vs the adult retina that was further reduced under hyperoxia. PRL and the PRL receptor mRNA were expressed at higher levels in the retina at P8 than in the adult, whereas endogenous PRL was undetectable in the circulation at P8. We conclude that PRL has a pro-angiogenic effect in the neonate retina as a result of its reduced conversion to vasoinhibin and that PRL produced by the retina may help promote physiological vascularisation after birth.
Collapse
Affiliation(s)
| | | | | | - Nundehui Díaz-Lezama
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Elva Adán-Castro
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | | | - Norma Adán
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | | | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| |
Collapse
|
19
|
Treatment of rams with melatonin implants in the non-breeding season improves post-thaw sperm progressive motility and DNA integrity. Anim Reprod Sci 2020; 221:106579. [PMID: 32919308 DOI: 10.1016/j.anireprosci.2020.106579] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 11/24/2022]
Abstract
In the Merino ram, it is unclear whether cryopreserved sperm function and fertility is compromised when collected during the non-breeding season, when Merino ewes are seasonally anestrus. It was therefore investigated whether treatment with melatonin could improve sperm function or fertility when semen was collected during the period Merino ewes were seasonally anestrus. There were 16 Merino rams treated or not treated with melatonin implants during the non-breeding season of ewes (September). Ejaculates were collected before melatonin treatment (Week 0), during the period of melatonin release (Week 7) and subsequent breeding season (Week 23). In vitro sperm function was assessed before freezing, and at 0- and 3 -hs post-thaw. Fertility was determined through intrauterine insemination of ewes (n = 966) with frozen-thawed samples, during the breeding season. Compared to Week 0 values, spermatozoa from melatonin-treated rams had greater progressive motility at Week 7 (P = 0.019) and less DNA fragmentation (P = 0.003) at Weeks 7 and 23, whilst spermatozoa from non-treated rams were unchanged during these time-periods. There were no other treatment effects on sperm function or fertility (P > 0.05). In ejaculates collected during Week 23, there were no effects of treatment either before freezing or post-thawing. Sperm from ejaculates collected at Week 23, however, had lesser pre-freezing/post-thawing total motility and resulted in lower pregnancy rates (P < 0.05). It is concluded there are no effects of season on sperm quality or fertility of Merino rams and that melatonin treatment subtly improves quality of spermatozoa following cryopreservation.
Collapse
|
20
|
Abstract
The hair cycle and hair follicle structure are highly affected by various hormones. Androgens—such as testosterone (T); dihydrotestosterone (DHT); and their prohormones, dehydroepiandrosterone sulfate (DHEAS) and androstendione (A)—are the key factors in terminal hair growth. They act on sex-specific areas of the body, converting small, straight, fair vellus hairs into larger darker terminal hairs. They bind to intracellular androgen receptors in the dermal papilla cells of the hair follicle. The majority of hair follicles also require the intracellular enzyme 5-alpha reductase to convert testosterone into DHT. Apart from androgens, the role of other hormones is also currently being researched—e.g., estradiol can significantly alter the hair follicle growth and cycle by binding to estrogen receptors and influencing aromatase activity, which is responsible for converting androgen into estrogen (E2). Progesterone, at the level of the hair follicle, decreases the conversion of testosterone into DHT. The influence of prolactin (PRL) on hair growth has also been intensively investigated, and PRL and PRL receptors were detected in human scalp skin. Our review includes results from many analyses and provides a comprehensive up-to-date understanding of the subject of the effects of hormonal changes on the hair follicle.
Collapse
|
21
|
Pool KR, Rickard JP, de Graaf SP. Overcoming neuroendocrine and metabolic barriers to puberty: the role of melatonin in advancing puberty in ewe lambs. Domest Anim Endocrinol 2020; 72:106457. [PMID: 32361422 DOI: 10.1016/j.domaniend.2020.106457] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/07/2020] [Accepted: 02/19/2020] [Indexed: 02/05/2023]
Abstract
Pubertal onset in the ewe is subject to a multitude of physiological and environmental constraints. As seasonal breeders, sheep rely on decreasing photoperiod to enter puberty and the subsequent breeding periods, hindering production. The initiation of puberty defines the reproductive yield of the ewe, and as such is a critical factor influencing production outcomes. Currently, the misconception that ovine puberty is reliant on age results in ewes being bred at over a year old, leading to a substantial unproductive period between birth and first conception. As such, transcending pubertal barriers to allow for earlier initiation of reproductive competency has significant commercial merit. The primary candidate to achieve this is the neurohormone melatonin, a key factor that naturally signals photoperiodic change that facilitates seasonal remodeling of the ovine hypothalamic-hypophyseal-gonadal axis. Despite being known to modulate reproductive seasonality in both the mature ewe and ram, the ability of melatonin to advance ewe puberty remains underutilized in industry. To optimize melatonin application and shape perceptions of breeding ewe lambs, a greater understanding of pubertal impediments and the natural role of melatonin is warranted. This review examines the physiological role and applications of melatonin to advance ewe puberty, and how this may act in conjunction with other physiological and metabolic cues.
Collapse
Affiliation(s)
- K R Pool
- The University of Sydney, Faculty of Science, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia.
| | - J P Rickard
- The University of Sydney, Faculty of Science, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
| | - S P de Graaf
- The University of Sydney, Faculty of Science, School of Life and Environmental Sciences, Sydney, NSW 2006, Australia
| |
Collapse
|
22
|
Trudeau VL, Somoza GM. Multimodal hypothalamo-hypophysial communication in the vertebrates. Gen Comp Endocrinol 2020; 293:113475. [PMID: 32240708 DOI: 10.1016/j.ygcen.2020.113475] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/21/2020] [Accepted: 03/28/2020] [Indexed: 12/13/2022]
Abstract
The vertebrate pituitary is arguably one of the most complex endocrine glands from the evolutionary, anatomical and functional perspectives. The pituitary plays a master role in endocrine physiology for the control of growth, metabolism, reproduction, water balance, and the stress response, among many other key processes. The synthesis and secretion of pituitary hormones are under the control of neurohormones produced by the hypothalamus. Under this conceptual framework, the communication between the hypophysiotropic brain and the pituitary gland is at the foundation of our understanding of endocrinology. The anatomy of the connections between the hypothalamus and the pituitary gland has been described in different vertebrate classes, revealing diverse modes of communication together with varying degrees of complexity. In this context, the evolution and variation in the neuronal, neurohemal, endocrine and paracrine modes will be reviewed in light of recent discoveries, and a re-evaluation of earlier observations. There appears to be three main hypothalamo-pituitary communication systems: 1. Diffusion, best exemplified by the agnathans; 2. Direct innervation of the adenohypophysis, which is most developed in teleost fish, and 3. The median eminence/portal blood vessel system, most conspicuously developed in tetrapods, showing also considerable variation between classes. Upon this basic classification, there exists various combinations possible, giving rise to taxon and species-specific, multimodal control over major physiological processes. Intrapituitary paracrine regulation and communication between folliculostellate cells and endocrine cells are additional processes of major importance. Thus, a more complex evolutionary picture of hypothalamo-hypophysial communication is emerging. There is currently little direct evidence to suggest which neuroendocrine genes may control the evolution of one communication system versus another. However, studies at the developmental and intergenerational timescales implicate several genes in the angiogenesis and axonal guidance pathways that may be important.
Collapse
Affiliation(s)
- Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada.
| | - Gustavo M Somoza
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Buenos Aires B7130IWA, Argentina.
| |
Collapse
|
23
|
Pool KR, Rickard JP, Pini T, de Graaf SP. Exogenous melatonin advances the ram breeding season and increases testicular function. Sci Rep 2020; 10:9711. [PMID: 32546776 PMCID: PMC7297710 DOI: 10.1038/s41598-020-66594-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/11/2020] [Indexed: 01/19/2023] Open
Abstract
Governed by melatonin, ovine reproductive seasonality limits production outcomes due to periods of decreased reproductive efficiency. Though it is established that slow-release melatonin implants improve out of season reproductive performance in the ewe, the comprehensive effects of exogenous melatonin in the ram remain inconclusive. This study aimed to ultimately clarify the ability of exogenous melatonin to alter ram reproductive function during the non-breeding season and the subsequent breeding season. Hence, we investigated the effect of exogenous melatonin on reproductive endocrinology, semen quality and production, testicular size and libido in Merino and Poll Dorset rams (n = 31, using a subset of 18 rams for analysis of semen production and quality). Melatonin treatment resulted in elevation of melatonin in seminal plasma from 1-8 weeks post-implantation and in blood plasma at 6 weeks post-implantation. The blood plasma testosterone of implanted rams was greater than controls at both 6 weeks post-implantation and during the following breeding season. Implanted rams exhibited increased testicular size and number of sperm per ejaculate from 3-12 weeks post-implantation but did not demonstrate any change in sperm motility or morphology in response to treatment. Compared to their control counterparts, melatonin-treated Poll Dorset rams exhibited a lower percentage of sperm DNA fragmentation during several weeks of the non-breeding season. Though melatonin increased the likelihood of ejaculate collection in Poll Dorset rams (P < 0.05), libido was otherwise unaffected by treatment. Melatonin did not alter seminal plasma concentrations of inhibin A or Anti-Mullerian hormone, however, for the first time in the ram we have shown Anti-Mullerian hormone to be positively correlated with the number of sperm per ejaculate and sperm motility (r = 0.464 and 0.3242 respectively, P < 0.001), and inhibin A to be correlated to the number of sperm per ejaculate (r = 0.1786, P = 0.0135). These results indicate that melatonin is able to both systemically upregulate reproduction and act directly upon testicular function in the ram.
Collapse
Affiliation(s)
- K R Pool
- The University of Sydney, Faculty of Science, School of Life and Environmental Sciences, Sydney, NSW, 2006, Australia.
| | - J P Rickard
- The University of Sydney, Faculty of Science, School of Life and Environmental Sciences, Sydney, NSW, 2006, Australia
| | - T Pini
- The University of Sydney, Faculty of Science, School of Life and Environmental Sciences, Sydney, NSW, 2006, Australia
| | - S P de Graaf
- The University of Sydney, Faculty of Science, School of Life and Environmental Sciences, Sydney, NSW, 2006, Australia
| |
Collapse
|
24
|
Dardente H, English WR, Valluru MK, Kanthou C, Simpson D. Debunking the Myth of the Endogenous Antiangiogenic Vegfaxxxb Transcripts. Trends Endocrinol Metab 2020; 31:398-409. [PMID: 32396842 DOI: 10.1016/j.tem.2020.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/28/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
In this opinion article we critically assess evidence for the existence of a family of antiangiogenic vascular endothelial growth factor (Vegfaxxxb) transcripts, arising from the use of a phylogenetically conserved alternative distal splice site within exon 8 of the VEGFA gene. We explain that prior evidence for Vegfaxxxb transcripts in tissues rests heavily upon flawed RT-PCR methodologies, with the extensive use of 5'-tailing in primer design being the main issue. Furthermore, our analysis of large RNA-seq data sets (human and ovine) fails to identify a single Vegfaxxxb transcript. Therefore, we challenge the very existence of Vegfaxxxb transcripts, which further questions the physiological relevance of studies based on the use of 'anti-VEGFAxxxb' antibodies. Our analysis has implications for the proposed therapeutic use of isoform-specific anti-VEGFA strategies for treating cancer and retinopathies.
Collapse
Affiliation(s)
- Hugues Dardente
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France.
| | - William R English
- Department of Oncology and Metabolism, Tumour Microcirculation Group, University of Sheffield, School of Medicine, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Manoj K Valluru
- Department of Oncology and Metabolism, Tumour Microcirculation Group, University of Sheffield, School of Medicine, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Chryso Kanthou
- Department of Oncology and Metabolism, Tumour Microcirculation Group, University of Sheffield, School of Medicine, Beech Hill Road, Sheffield, S10 2RX, UK
| | - David Simpson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT7 1NN, UK
| |
Collapse
|
25
|
Mirza-Aghazadeh-Attari M, Reiter RJ, Rikhtegar R, Jalili J, Hajalioghli P, Mihanfar A, Majidinia M, Yousefi B. Melatonin: An atypical hormone with major functions in the regulation of angiogenesis. IUBMB Life 2020; 72:1560-1584. [PMID: 32329956 DOI: 10.1002/iub.2287] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/10/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
Melatonin (N-acetyl-5-methoxytryptamine), a pleotropic molecule with a wide distribution, has received considerable attention in recent years, mostly because of its various major effects on tissues or cells since it has both receptor-dependent and receptor-independent actions over a wide range of concentrations. These biological and physiological functions of melatonin include regulation of circadian rhythms by modulating the expression of core oscillator genes, scavenging the reactive oxygen species and reactive nitrogen species, modulating the immune system and inflammatory response, and exerting cytoprotective and antiapoptotic effects. Given the multiple critical roles of melatonin, dysregulation of its production or any disruption in signaling through its receptors may have contributed in the development of a wide range of disorders including type 2 diabetes, aging, immune-mediated diseases, hypertension, and cancer. Herein, we focus on the modulatory effects of melatonin on angiogenesis and its implications as a therapeutic strategy in cancer and related diseases.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Reza Rikhtegar
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Jalili
- Radiology Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hajalioghli
- Radiology Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ainaz Mihanfar
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Ma Q, Reiter RJ, Chen Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis 2019; 23:91-104. [PMID: 31650428 DOI: 10.1007/s10456-019-09689-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/09/2019] [Indexed: 02/07/2023]
Abstract
Angiogenesis depends on proangiogenic and anti-angiogenic molecules that regulate endothelial cell proliferation and migration. Well-regulated angiogenesis plays a pivotal role in many physiological conditions such as reproduction and embryonic development, while abnormal angiogenesis is also the basis of a variety of pathological processes including tumor metastasis and atherosclerotic plaque formation. Melatonin has a variety of biological effects, including inhibition of tumor metastasis, stabilizing atherosclerotic plaques, and the regulation of seasonal reproductive rhythms, etc. During certain pathophysiological processes, melatonin exerts different functions depending on its ability to regulate angiogenesis. This review reveals that melatonin has different effects on neovascularization under different physiological and pathological conditions. In tumors, in age-related ocular diseases, and in a hypoxic environment, melatonin inhibits neovascularization in tissues, while in gastric ulcers, skin lesions, and some physiologic processes, it promotes angiogenesis. We also speculate that melatonin may inhibit the neovascularization in atherosclerotic plaques, thus preventing the initiation and development of atherosclerosis. Most studies suggest that these effects are related to the role of melatonin in regulating of vascular endothelial growth factor and its receptors, but the specific regulatory mechanisms remain disparate, which may lead to the differential effects of melatonin on angiogenesis under different conditions. In this review, we thus summarize some seemingly contradictory mechanisms by which melatonin controls angiogenesis under different pathological and physiological conditions, and urge that the regulatory mechanisms be further studied.
Collapse
Affiliation(s)
- Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, Texas, 78229, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, Texas, 78229, USA.
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
27
|
Kandemir YB, Konuk E, Katırcı E, Xxx F, Behram M. Is the effect of melatonin on vascular endothelial growth factor receptor-2 associated with angiogenesis in the rat ovary? Clinics (Sao Paulo) 2019; 74:e658. [PMID: 30864638 PMCID: PMC6438131 DOI: 10.6061/clinics/2019/e658] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Vascular endothelial growth factor (VEGF) and its receptors play important roles in angiogenesis. Melatonin plays an important role in gonadal development; thus, its effect on the reproductive system is evident. We investigated the influence of melatonin on the expression of VEGF, vascular endothelial growth factor receptor-1 (VEGFR1) and vascular endothelial growth factor receptor-2 (VEGFR2), as well as on changes in oxidative stress markers and follicle numbers in rat ovaries. METHODS For this purpose, 45 Wistar rats were separated into the following groups: Group 1, control; Group 2, vehicle; and Group 3, melatonin. Rats in Group 3 were treated with melatonin at 50 mg/kg/day for 30 days. The effects of melatonin on the expression of VEGF, VEGFR1 and VEGFR2 were established by immunohistochemistry analysis. The effects of melatonin on antioxidant enzyme activities were demonstrated by spectrophotometric analysis. RESULTS Based on immunohistochemistry analysis, VEGFR2 was predominantly localized to theca cells in the ovary. Our data indicate that melatonin treatment can significantly increase VEGF and VEGFR1 expression in the ovary ( p <0.05). Additionally, the number of degenerated follicles significantly decreased with melatonin treatment ( p <0.05). Melatonin administration also led to significant increases in antioxidant enzyme levels in the ovary. CONCLUSION Melatonin treatment exerts protective effects on follicles against increased lipid peroxidation through modulating tissue antioxidant enzyme levels. These effects may be related to angiogenesis and antioxidant activities.
Collapse
Affiliation(s)
- Yasemin Behram Kandemir
- Harran University, Faculty of Medicine, Department of Anatomy, Şanlıurfa, Turkey
- Corresponding author. E-mail:
| | - Esma Konuk
- Akdeniz University, Faculty of Medicine, Department of Histology, Antalya, Turkey
| | - Ertan Katırcı
- Akdeniz University, Faculty of Medicine, Department of Histology, Antalya, Turkey
| | - Feride Xxx
- Akdeniz University, Faculty of Medicine, Department of Histology, Antalya, Turkey
| | - Mustafa Behram
- Kanuni Sultan Süleyman Hospital, Department of Perinatology, Istanbul, Turkey
| |
Collapse
|
28
|
Lincoln G. A brief history of circannual time. J Neuroendocrinol 2019; 31:e12694. [PMID: 30739343 DOI: 10.1111/jne.12694] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/31/2019] [Accepted: 02/05/2019] [Indexed: 12/22/2022]
Abstract
Innate circannual timing is an ancestral trait that first evolved in free-living eukaryotic cells some 2000 million years ago, with marine algae of the genus Allexandrium providing a living unicellular model. This species shows the primitive trait of 'alternation of generations', where the organism alternates between fast replicating vegetative cells in the summer and a dormant cystic cell over the winter. The resistant cysts sink into the cold ocean sediments. Remarkably, excystment in spring is governed by an endogenous circannual timing mechanism. Thus, a tiny, short-lived unicell can utilise a circannual clock as part of the life-history programme of the species. Innate timing allows for major adjustments in physiology and behaviour in anticipation of the seasons, and provides an internalised sense of seasonal time for the many species where standard environmental cues are weak or ambiguous. This is a highly adaptive strategy irrespective of the size and longevity of an organism. Circannual rhythms are expressed by a diverse range of organisms, from flowering plants to mammals, interwoven into the life-history programme of each species, being a consequence of forever living in a periodic world. In complex vertebrates, the early division of the zygote potentially carries circannual timer genes into all progeny cells and tissues. This supports the concept of a 'clock-shop' where cell-autonomous long-term rhythms are generated in each tissue, orchestrated by a central circannual pacemaker system. This is analogous to the organisation of the circadian timing system. For the circannual time-scale, specialised thyrotroph cells located in the pars tuberalis of the pituitary gland and adjacent tanycyte cells located in the ependymal wall of the third cerebral ventricle of the brain act as putative central circannual pacemakers. At a molecular level, epigenetically regulated, cyclical remodelling of chromatin, which determines whether specific circannual timer genes are transcriptionally active, or not, is considered to drive the oscillation between the summer and winter phenotypes.
Collapse
|
29
|
Dusart P, Fagerberg L, Perisic L, Civelek M, Struck E, Hedin U, Uhlén M, Trégouët DA, Renné T, Odeberg J, Butler LM. A systems-approach reveals human nestin is an endothelial-enriched, angiogenesis-independent intermediate filament protein. Sci Rep 2018; 8:14668. [PMID: 30279450 PMCID: PMC6168570 DOI: 10.1038/s41598-018-32859-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/12/2018] [Indexed: 12/18/2022] Open
Abstract
The intermediate filament protein nestin is expressed during embryonic development, but considered largely restricted to areas of regeneration in the adult. Here, we perform a body-wide transcriptome and protein-profiling analysis to reveal that nestin is constitutively, and highly-selectively, expressed in adult human endothelial cells (EC), independent of proliferative status. Correspondingly, we demonstrate that it is not a marker for tumour EC in multiple malignancy types. Imaging of EC from different vascular beds reveals nestin subcellular distribution is shear-modulated. siRNA inhibition of nestin increases EC proliferation, and nestin expression is reduced in atherosclerotic plaque neovessels. eQTL analysis reveals an association between SNPs linked to cardiovascular disease and reduced aortic EC nestin mRNA expression. Our study challenges the dogma that nestin is a marker of proliferation, and provides insight into its regulation and function in EC. Furthermore, our systems-based approach can be applied to investigate body-wide expression profiles of any candidate protein.
Collapse
Affiliation(s)
- Philip Dusart
- Science for Life Laboratory, School of Biotechnology, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, School of Biotechnology, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Ljubica Perisic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, SE-171 76, Stockholm, Sweden
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia, Charlottesville, USA
| | - Eike Struck
- Science for Life Laboratory, School of Biotechnology, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - Ulf Hedin
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, SE-171 76, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, School of Biotechnology, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, SE-171 21, Stockholm, Sweden
| | - David-Alexandre Trégouët
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France.,ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Thomas Renné
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, D-20246, Hamburg, Germany
| | - Jacob Odeberg
- Science for Life Laboratory, School of Biotechnology, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, SE-171 21, Stockholm, Sweden.,Coagulation Unit, Centre for Hematology, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Lynn M Butler
- Science for Life Laboratory, School of Biotechnology, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, SE-171 21, Stockholm, Sweden. .,Clinical Chemistry and Blood Coagulation, Department of Molecular Medicine and Surgery, Karolinska Institute, SE-171 76, Stockholm, Sweden. .,Institute for Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, D-20246, Hamburg, Germany.
| |
Collapse
|
30
|
Nie JJ, Qiao B, Duan S, Xu C, Chen B, Hao W, Yu B, Li Y, Du J, Xu FJ. Unlockable Nanocomplexes with Self-Accelerating Nucleic Acid Release for Effective Staged Gene Therapy of Cardiovascular Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801570. [PMID: 29920798 DOI: 10.1002/adma.201801570] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 04/26/2018] [Indexed: 05/24/2023]
Abstract
Nucleic acid (NA)-based therapy is proposed to address serious diseases such as cardiovascular diseases (CVDs). Powerful NA delivery vehicles are essential for effective gene therapy. Herein, a novel type of delivery vehicle, an unlockable core-shell nanocomplex (Hep@PGEA) with self-accelerating NA release, is structurally designed. Hep@PGEA is composed of disulfide-bridged heparin nanoparticle (HepNP) core and low-toxicity PGEA cationic shell. In comparison with NA, heparin, a negatively charged polysaccharide macromolecule, exhibits stronger interactions with cationic species. Upon the breakdown of redox-responsive HepNP cores, unlocked heparin would interact with the outer cationic shells and replace the condensed NA to facilitate NA release. Such unique Hep@PGEA is successfully explored for effective miRNA-pDNA staged gene therapy of myocardial infarction (MI), one of the most serious CVDs. With the progression of MI, glutathione amounts in heart tissues increase. MiR-499 (for the inhibition of cardiomyocyte apoptosis) and plasmid encoding vascular endothelial growth factor (for the promotion of angiogenesis) are sequentially delivered for systemic treatment of MI. Such treatment produces impressive results in restoring heart function and suppressing cardiac hypertrophy. Due to the wide existence of redox agents in cells, the proposed unlockable delivery nanovehicle and staged therapy strategy can provide new methods to effectively treat different serious diseases.
Collapse
Affiliation(s)
- Jing-Jun Nie
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Bokang Qiao
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), and Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Shun Duan
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chen Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Boya Chen
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), and Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Wenjing Hao
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), and Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Bingran Yu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yulin Li
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), and Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Jie Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), and Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, China
| | - Fu-Jian Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
31
|
Tabecka-Lonczynska A, Mytych J, Solek P, Kulpa-Greszta M, Sowa-Kucma M, Koziorowski M. Vascular endothelial growth factor (VEGF-A) and fibroblast growth factor (FGF-2) as potential regulators of seasonal reproductive processes in male European bison (Bison bonasus, Linnaeus 1758). Gen Comp Endocrinol 2018; 263:72-79. [PMID: 29626449 DOI: 10.1016/j.ygcen.2018.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 03/04/2018] [Accepted: 04/03/2018] [Indexed: 11/25/2022]
Abstract
Growth factors: vascular endothelial growth factor A (VEGF-A) and fibroblast growth factor (FGF-2) were reported to affect normal physiological reproductive processes in human, domestic and free living animals. Moreover, some reports suggest that VEGF-A and FGF-2 may be directly involved in the control of the annual reproductive cycle of seasonally breeding animals but detailed knowledge is still missing. Our study aimed to demonstrate the expression of mRNA and protein for both factors in the tissues of testis and epididymis (caput, corpus, cauda) at different periods of the year (March, June, November, December) in European bison as a model of seasonally breeding animal. Results suggest, that VEGF-A expression was more pronounced in testis than in epididymis and the highest expression was noted in December and June. Surprisingly, the highest protein accumulation was observed in June at the same level in all tissues analyzed. On the other hand, the highest FGF-2 mRNA expression was noted in testis in June and in epididymis in March. However, no differences in protein expression of FGF-2 were found between analyzed groups. The results indicate that both factors are necessary for proper functioning of the reproductive system and their levels differ seasonally. Perhaps, it is linked to increased need of these factors in the testis as well as epididymis during preparation for the reproductive functions. Moreover, VEGF-A and FGF-2 not only may regulate reproductive functions by affecting vascularization and cell nutrition, but it also may be possible that they possess protective functions by stabilizing the reproductive cells. Therefore, obtained results provide new insight into mechanisms underlying seasonal breeding of the male European bison.
Collapse
Affiliation(s)
- Anna Tabecka-Lonczynska
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland.
| | - Jennifer Mytych
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| | - Przemyslaw Solek
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| | - Magdalena Kulpa-Greszta
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| | - Magdalena Sowa-Kucma
- Institute of Pharmacology, Polish Academy of Sciences, Department of Neurobiology, Laboratory of Trace Elements Neurobiology, Smeta 12, 31-343 Kraków, Poland; Department of Human Physiology, Institute of Clinical and Experimental Medicine, Medical Faculty, University of Rzeszow, Kopisto 2a, 35-310 Rzeszów, Poland
| | - Marek Koziorowski
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| |
Collapse
|
32
|
Bates DO, Beazley-Long N, Benest AV, Ye X, Ved N, Hulse RP, Barratt S, Machado MJ, Donaldson LF, Harper SJ, Peiris-Pages M, Tortonese DJ, Oltean S, Foster RR. Physiological Role of Vascular Endothelial Growth Factors as Homeostatic Regulators. Compr Physiol 2018; 8:955-979. [PMID: 29978898 DOI: 10.1002/cphy.c170015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The vascular endothelial growth factor (VEGF) family of proteins are key regulators of physiological systems. Originally linked with endothelial function, they have since become understood to be principal regulators of multiple tissues, both through their actions on vascular cells, but also through direct actions on other tissue types, including epithelial cells, neurons, and the immune system. The complexity of the five members of the gene family in terms of their different splice isoforms, differential translation, and specific localizations have enabled tissues to use these potent signaling molecules to control how they function to maintain their environment. This homeostatic function of VEGFs has been less intensely studied than their involvement in disease processes, development, and reproduction, but they still play a substantial and significant role in healthy control of blood volume and pressure, interstitial volume and drainage, renal and lung function, immunity, and signal processing in the peripheral and central nervous system. The widespread expression of VEGFs in healthy adult tissues, and the disturbances seen when VEGF signaling is inhibited support this view of the proteins as endogenous regulators of normal physiological function. This review summarizes the evidence and recent breakthroughs in understanding of the physiology that is regulated by VEGF, with emphasis on the role they play in maintaining homeostasis. © 2017 American Physiological Society. Compr Physiol 8:955-979, 2018.
Collapse
Affiliation(s)
- David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | | | - Andrew V Benest
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Xi Ye
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Nikita Ved
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Richard P Hulse
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Shaney Barratt
- Academic Respiratory Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Maria J Machado
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Lucy F Donaldson
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Steven J Harper
- School of Physiology, Pharmacology & Neuroscience, Medical School, University of Bristol, Bristol, United Kingdom
| | - Maria Peiris-Pages
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Domingo J Tortonese
- Centre for Comparative and Clinical Anatomy, University of Bristol, Bristol, United Kingdom
| | - Sebastian Oltean
- Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Rebecca R Foster
- Bristol Renal, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
33
|
Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism. Endocr Rev 2018; 39:333-368. [PMID: 29351662 DOI: 10.1210/er.2017-00235] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
The fertility and survival of an individual rely on the ability of the periphery to promptly, effectively, and reproducibly communicate with brain neural networks that control reproduction, food intake, and energy homeostasis. Tanycytes, a specialized glial cell type lining the wall of the third ventricle in the median eminence of the hypothalamus, appear to act as the linchpin of these processes by dynamically controlling the secretion of neuropeptides into the portal vasculature by hypothalamic neurons and regulating blood-brain and blood-cerebrospinal fluid exchanges, both processes that depend on the ability of these cells to adapt their morphology to the physiological state of the individual. In addition to their barrier properties, tanycytes possess the ability to sense blood glucose levels, and play a fundamental and active role in shuttling circulating metabolic signals to hypothalamic neurons that control food intake. Moreover, accumulating data suggest that, in keeping with their putative descent from radial glial cells, tanycytes are endowed with neural stem cell properties and may respond to dietary or reproductive cues by modulating hypothalamic neurogenesis. Tanycytes could thus constitute the missing link in the loop connecting behavior, hormonal changes, signal transduction, central neuronal activation and, finally, behavior again. In this article, we will examine these recent advances in the understanding of tanycytic plasticity and function in the hypothalamus and the underlying molecular mechanisms. We will also discuss the putative involvement and therapeutic potential of hypothalamic tanycytes in metabolic and fertility disorders.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Bénédicte Dehouck
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Ariane Sharif
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Philippe Ciofi
- Inserm, Neurocentre Magendie, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| |
Collapse
|
34
|
Anti-angiogenic VEGFAxxxb transcripts are not expressed in the medio-basal hypothalamus of the seasonal sheep. PLoS One 2018; 13:e0197123. [PMID: 29746548 PMCID: PMC5944957 DOI: 10.1371/journal.pone.0197123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 04/25/2018] [Indexed: 12/15/2022] Open
Abstract
This study investigated Vegfa expression in the pars tuberalis (PT) of the pituitary and medio-basal hypothalamus (MBH) of sheep, across seasons and reproductive states. It has recently been proposed that season impacts alternative splicing of Vegfa mRNA in the PT, which shifts the balance between angiogenic VEGFAxxx and anti-angiogenic VEGFAxxxb isoforms (with xxx the number of amino acids of the mature VEGFA proteins) to modulate seasonal breeding. Here, we used various RT-PCR methodologies and analysis of RNAseq datasets to investigate seasonal variation in expression and splicing of the ovine Vegfa gene. Collectively, we identify 5 different transcripts for Vegfa within the ewe PT/MBH, which correspond to splicing events previously described in mouse and human. All identified transcripts encode angiogenic VEGFAxxx isoforms, with no evidence for alternative splicing within exon 8. These findings led us to investigate in detail how "Vegfaxxxb-like" PCR products could be generated by RT-PCR and misidentified as endogenous transcripts, in sheep and human HEK293 cells. In conclusion, our findings do not support the existence of anti-angiogenic VEGFAxxxb isoforms in the ovine PT/MBH and shed new light on the interpretation of prior studies, which claimed to identify Vegfaxxxb isoforms by RT-PCR.
Collapse
|
35
|
Abstract
The circumventricular organs (CVOs) are specialised neuroepithelial structures found in the midline of the brain, grouped around the third and fourth ventricles. They mediate the communication between the brain and the periphery by performing sensory and secretory roles, facilitated by increased vascularisation and the absence of a blood-brain barrier. Surprisingly little is known about the origins of the CVOs (both developmental and evolutionary), but their functional and organisational similarities raise the question of the extent of their relationship. Here, I review our current knowledge of the embryonic development of the seven major CVOs (area postrema, median eminence, neurohypophysis, organum vasculosum of the lamina terminalis, pineal organ, subcommissural organ, subfornical organ) in embryos of different vertebrate species. Although there are conspicuous similarities between subsets of CVOs, no unifying feature characteristic of their development has been identified. Cross-species comparisons suggest that CVOs also display a high degree of evolutionary flexibility. Thus, the term 'CVO' is merely a functional definition, and features shared by multiple CVOs may be the result of homoplasy rather than ontogenetic or phylogenetic relationships.
Collapse
Affiliation(s)
- Clemens Kiecker
- Department of Developmental NeurobiologyKing's College LondonLondonUK
| |
Collapse
|
36
|
Korf HW. Signaling pathways to and from the hypophysial pars tuberalis, an important center for the control of seasonal rhythms. Gen Comp Endocrinol 2018; 258:236-243. [PMID: 28511899 DOI: 10.1016/j.ygcen.2017.05.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 11/28/2022]
Abstract
Seasonal (circannual) rhythms play an important role for the control of body functions (reproduction, metabolism, immune responses) in nearly all living organisms. Also humans are affected by the seasons with regard to immune responses and mental functions, the seasonal affective disorder being one of the most prominent examples. The hypophysial pars tuberalis (PT), an important interface between the hypophysial pars distalis and neuroendocrine centers in the brain, plays an essential role in the regulation of seasonal functions and may even be the seat of the circannual clock. Photoperiodic signals provide a major input to the PT. While the perception of these signals involves extraocular photoreceptors in non-mammalian species (birds, fish), mammals perceive photoperiodic signals exclusively in the retina. A multisynaptic pathway connects the retina with the pineal organ where photoperiodic signals are translated into the neurohormone melatonin that is rhythmically produced night by night and encodes the length of the night. Melatonin controls the functional activity of the mammalian PT by acting upon MT1 melatonin receptors. The PT sends its output signals via retrograde and anterograde pathways. The retrograde pathway targetting the hypothalamus employs TSH as messenger and controls a local hypothalamic T3 system. As discovered in Japanese quail, TSH triggers molecular cascades mediating thyroid hormone conversion in the ependymal cell layer of the infundibular recess of the third ventricle. The local accumulation of T3 in the mediobasal hypothalamus (MBH) appears to activate the gonadal axis by affecting the neuro-glial interaction between GnRH terminals and tanycytes in the median eminence. This retrograde pathway is conserved in photoperiodic mammals (sheep and hamsters), and even in non-photoperiodic laboratory mice provided that they are capable to synthesize melatonin. The anterograde pathway is implicated in the control of prolactin secretion, targets cells in the PD and supposedly employs small molecules as signal substances collectively denominated as "tuberalins". Several "tuberalin" candidates have been proposed, such as tachykinins, the secretory protein TAFA and endocannabinoids (EC). The PT-intrinsic EC system was first demonstrated in Syrian hamsters and shown to respond to photoperiodic changes. Subsequently, the EC system was also demonstrated in the PT of mice, rats and humans. To date, 2-arachidonoylglycerol (2-AG) appears as the most important endocannabinoid from the PT. Likely targets for the EC are folliculo-stellate cells that contain the CB1 receptor and appear to contact lactotroph cells. The CB1 receptor was also found on corticotroph cells which appear as a further target of the EC. Recently, the CB1 receptor was also localized to CRF-containing nerve fibers running in the outer zone of the median eminence. This finding suggests that the EC system of the PT contributes not only to the anterograde, but also to the retrograde pathway. Taken together, the results support the concept that the PT transmits its signals via a "cocktail" of messenger molecules which operate also in other brain areas and systems rather than through PT-specific "tuberalins". Furthermore, they may attribute a novel function to the PT, namely the modulation of the stress response and immune functions.
Collapse
Affiliation(s)
- Horst-Werner Korf
- Dr. Senckenbergische Anatomie, Institut für Anatomie II, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
37
|
Huxley VH, Kemp SS. Sex-Specific Characteristics of the Microcirculation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:307-328. [PMID: 30051393 DOI: 10.1007/978-3-319-77932-4_20] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The requirements of metabolizing tissue are both continuous and variable; accordingly, the microvasculature serving that tissue must be similarly dynamic. Just as it is recognized that males and females of the same species have differing metabolic requirements, is it not likely that the microvasculature serving these tissues will differ by sex? This section focusing on the constituents of the microcirculation identifies what is known presently about the role sex plays in matching metabolic demand with microvascular function and areas requiring additional study. Many of the identified sex differences are subtle and easily ignored. In the aggregate, though, they can profoundly alter phenotype, especially under stressful conditions including pregnancy, exercise, and disease states ranging from diabetes to heart failure. Although the features presently identified to "have sex" range from differences in growth, morphology, protein expression, and intracellular signaling, males and females alike achieve homeostasis, likely by different means. Studies of microvascular sexual dimorphism are also identifying age as an independent but interacting factor requiring additional attention. Overall, attempting to ignore either sex and/or age is inappropriate and will prevent the design and implementation of appropriate interventions to present, ameliorate, or correct microvascular dysfunction.
Collapse
Affiliation(s)
- Virginia H Huxley
- Center for Gender Physiology, Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA.
| | - Scott S Kemp
- Center for Gender Physiology, Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
38
|
Melatonin and Fertoprotective Adjuvants: Prevention against Premature Ovarian Failure during Chemotherapy. Int J Mol Sci 2017; 18:ijms18061221. [PMID: 28590419 PMCID: PMC5486044 DOI: 10.3390/ijms18061221] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/02/2017] [Accepted: 06/05/2017] [Indexed: 12/22/2022] Open
Abstract
Premature ovarian failure is one of the side effects of chemotherapy in pre-menopausal cancer patients. Preservation of fertility has become increasingly important in improving the quality of life of completely recovered cancer patients. Among the possible strategies for preserving fertility such as ovarian tissue cryopreservation, co-treatment with a pharmacological adjuvant is highly effective and poses less of a burden on the human body. Melatonin is generally produced in various tissues and acts as a universally acting antioxidant in cells. Melatonin is now more widely used in various biological processes including treating insomnia and an adjuvant during chemotherapy. In this review, we summarize the information indicating that melatonin may be useful for reducing and preventing premature ovarian failure in chemotherapy-treated female patients. We also mention that many adjuvants other than melatonin are developed and used to inhibit chemotherapy-induced infertility. This information will give us novel insights on the clinical use of melatonin and other agents as fertoprotective adjuvants for female cancer patients.
Collapse
|