1
|
Iversen JN, Tai YK, Wu KY, Wong CJK, Lim HY, Franco-Obregón A. Magnetically Stimulated Myogenesis Recruits a CRY2-TRPC1 Photosensitive Signaling Axis. Cells 2025; 14:231. [PMID: 39937022 PMCID: PMC11817702 DOI: 10.3390/cells14030231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
The cryptochromes are flavoproteins that either individually or synergistically respond to light and magnetic field directionality as well as are implicated in circadian rhythm entrainment and development. Single brief exposures (10 min) to low energy (1.5 mT) pulsed electromagnetic fields (PEMFs) were previously shown to enhance myogenesis by stimulating transient receptor potential canonical 1 (TRPC1)-mediated Ca2+ entry, whereby downwardly directed fields produced greater myogenic enhancement than upwardly directed fields. Here, we show that growth in the dark results in myoblasts losing their sensitivity to both magnetic field exposure and directionality. By contrast, overexpressing or silencing cryptochrome circadian regulator 2 (CRY2) in myoblasts enhances or reduces PEMF responses, respectively, under conditions of ambient light. Reducing cellular flavin adenine dinucleotide (FAD) content by silencing riboflavin kinase (RFK) attenuated responsiveness to PEMFs and inhibited selectivity for magnetic field direction. The upregulation of TRPC1 and cell cycle regulatory proteins typically observed in response to PEMF exposure was instead attenuated by upwardly directed magnetic fields, growth in the darkness, magnetic shielding, or the silencing of CRY2 or RFK. A physical interaction between CRY2 and TRPC1 was detected using coimmunoprecipitation and immunofluorescence, revealing their co-translocation into the nucleus after PEMF exposure. These results implicate CRY2 in an identified TRPC1-dependent magnetotransduction myogenic cascade.
Collapse
Affiliation(s)
- Jan Nikolas Iversen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (K.Y.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore;
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (K.Y.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore;
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Kwan Yu Wu
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (K.Y.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore;
| | - Craig Jun Kit Wong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (K.Y.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore;
| | - Hao Yang Lim
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore;
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (K.Y.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore;
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
2
|
Zou XF, Zhang BZ, Qian WW, Cheng FM. Bone marrow mesenchymal stem cells in treatment of peripheral nerve injury. World J Stem Cells 2024; 16:799-810. [PMID: 39219723 PMCID: PMC11362854 DOI: 10.4252/wjsc.v16.i8.799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/20/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
Peripheral nerve injury (PNI) is a common neurological disorder and complete functional recovery is difficult to achieve. In recent years, bone marrow mesenchymal stem cells (BMSCs) have emerged as ideal seed cells for PNI treatment due to their strong differentiation potential and autologous transplantation ability. This review aims to summarize the molecular mechanisms by which BMSCs mediate nerve repair in PNI. The key mechanisms discussed include the differentiation of BMSCs into multiple types of nerve cells to promote repair of nerve injury. BMSCs also create a microenvironment suitable for neuronal survival and regeneration through the secretion of neurotrophic factors, extracellular matrix molecules, and adhesion molecules. Additionally, BMSCs release pro-angiogenic factors to promote the formation of new blood vessels. They modulate cytokine expression and regulate macrophage polarization, leading to immunomodulation. Furthermore, BMSCs synthesize and release proteins related to myelin sheath formation and axonal regeneration, thereby promoting neuronal repair and regeneration. Moreover, this review explores methods of applying BMSCs in PNI treatment, including direct cell transplantation into the injured neural tissue, implantation of BMSCs into nerve conduits providing support, and the application of genetically modified BMSCs, among others. These findings confirm the potential of BMSCs in treating PNI. However, with the development of this field, it is crucial to address issues related to BMSC therapy, including establishing standards for extracting, identifying, and cultivating BMSCs, as well as selecting application methods for BMSCs in PNI such as direct transplantation, tissue engineering, and genetic engineering. Addressing these issues will help translate current preclinical research results into clinical practice, providing new and effective treatment strategies for patients with PNI.
Collapse
Affiliation(s)
- Xiong-Fei Zou
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Bao-Zhong Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing 100730, China.
| | - Wen-Wei Qian
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Florence Mei Cheng
- College of Nursing, The Ohio State University, Ohio, OH 43210, United States
| |
Collapse
|
3
|
Ritter A, Tessmar-Raible K. Time me by the moon : The evolution and function of lunar timing systems. EMBO Rep 2024; 25:3169-3176. [PMID: 39014253 PMCID: PMC11316100 DOI: 10.1038/s44319-024-00196-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024] Open
Abstract
The moon has significant impact on the timing of organisms. Can the study of molecular timing mechanisms of marine animals and algae help to understand some of the “weird” correlations between human physiological/behavioral rhythms and the lunar cycle?
Collapse
Affiliation(s)
- Andrés Ritter
- Laboratory of Integrative Biology of Marine Models, UMR8227 Sorbonne Université-CNRS, Station Biologique de Roscoff, 29688, Roscoff, CEDEX, France.
| | - Kristin Tessmar-Raible
- Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, 1030, Vienna, Austria.
- Alfred Wegener Institute Helmholtz Centre for Polar and Marine Research, Am Handelshafen 12, 27570, Bremerhaven, Germany.
- Institute for Chemistry and Biology of the Marine Environment (ICBM), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129, Oldenburg, Germany.
| |
Collapse
|
4
|
Parasram K, Zuccato A, Shin M, Willms R, DeVeale B, Foley E, Karpowicz P. The emergence of circadian timekeeping in the intestine. Nat Commun 2024; 15:1788. [PMID: 38413599 PMCID: PMC10899604 DOI: 10.1038/s41467-024-45942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
The circadian clock is a molecular timekeeper, present from cyanobacteria to mammals, that coordinates internal physiology with the external environment. The clock has a 24-h period however development proceeds with its own timing, raising the question of how these interact. Using the intestine of Drosophila melanogaster as a model for organ development, we track how and when the circadian clock emerges in specific cell types. We find that the circadian clock begins abruptly in the adult intestine and gradually synchronizes to the environment after intestinal development is complete. This delayed start occurs because individual cells at earlier stages lack the complete circadian clock gene network. As the intestine develops, the circadian clock is first consolidated in intestinal stem cells with changes in Ecdysone and Hnf4 signalling influencing the transcriptional activity of Clk/cyc to drive the expression of tim, Pdp1, and vri. In the mature intestine, stem cell lineage commitment transiently disrupts clock activity in differentiating progeny, mirroring early developmental clock-less transitions. Our data show that clock function and differentiation are incompatible and provide a paradigm for studying circadian clocks in development and stem cell lineages.
Collapse
Affiliation(s)
- Kathyani Parasram
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Amy Zuccato
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Minjeong Shin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Reegan Willms
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Brian DeVeale
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Phillip Karpowicz
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada.
| |
Collapse
|
5
|
Abdelbaset-Ismail A, Brzezniakiewicz-Janus K, Thapa A, Ratajczak J, Kucia M, Ratajczak MZ. Pineal Gland Hormone Melatonin Inhibits Migration of Hematopoietic Stem/Progenitor Cells (HSPCs) by Downregulating Nlrp3 Inflammasome and Upregulating Heme Oxygenase-1 (HO-1) Activity. Stem Cell Rev Rep 2024; 20:237-246. [PMID: 37812364 DOI: 10.1007/s12015-023-10638-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/10/2023]
Abstract
Hematopoietic stem progenitor cells (HSPCs) follow the diurnal circulation rhythm in peripheral blood (PB) with nadir during late night and peak at early morning hours. The level of these cells in PB correlates with activation of innate immunity pathways, including complement cascade (ComC) that drives activation of Nlrp3 inflammasome. To support this, mice both in defective ComC activation as well as Nlrp3 inflammasome do not show typical changes in the diurnal level of circulating HSPCs. Migration of HSPCs is also impaired at the intracellular level by the anti-inflammatory enzyme heme oxygenase-1 (HO-1) which is an inhibitor of Nlrp3 inflammasome. It is also well known that circadian rhythm mediates PB level of melatonin released from the pineal gland. Since trafficking of HSPCs is driven by innate immunity-induced sterile inflammation and melatonin has an anti-inflammatory effect, we hypothesized that melatonin could negatively impact the release of HSPCs from BM into PB by inhibiting Nlrp3 inflammasome activation. We provide an evidence that melatonin being a ''sleep regulating pineal hormone'' directly inhibits migration of HSPCs both in vitro migration assays and in vivo during pharmacological mobilization. This correlated with inhibition of cholesterol synthesis required for a proper membrane lipid raft (MLRs) formation and an increase in expression of HO-1-an inhibitor of Nlrp3 inflammasome. Since melatonin is a commonly used drug, this should be considered while preparing a patient for the procedure of HSPCs mobilization. More importantly, our studies shed more mechanistic light on a role of melatonin in the diurnal circulation of HSPCs.
Collapse
Affiliation(s)
- Ahmed Abdelbaset-Ismail
- Stem Cell Institute at Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
- Surgery, Anesthesiology, and Radiology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | | | - Arjun Thapa
- Stem Cell Institute at Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Janina Ratajczak
- Stem Cell Institute at Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Magda Kucia
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Institute at Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA.
- Department of Hematology, University of Zielona Gora, Multi-Specialist Hospital Gorzow Wlkp., Gorzow Wlkp., Poland.
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
6
|
Hearn JI, Alhilali M, Kim M, Kalev-Zylinska ML, Poulsen RC. N-methyl-D-aspartate receptor regulates the circadian clock in megakaryocytic cells and impacts cell proliferation through BMAL1. Platelets 2023; 34:2206918. [PMID: 37183795 DOI: 10.1080/09537104.2023.2206918] [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: 08/13/2022] [Revised: 02/16/2023] [Accepted: 04/17/2023] [Indexed: 05/16/2023]
Abstract
Peripheral circadian clocks control cell proliferation and survival, but little is known about their role and regulation in megakaryocytic cells. N-methyl-D-aspartate receptor (NMDAR) regulates the central clock in the brain. The purpose of this study was to determine whether NMDAR regulates the megakaryocytic cell clock and whether the megakaryocytic clock regulates cell proliferation and cell death. We found that both the Meg-01 megakaryocytic cell line and native murine megakaryocytes expressed circadian clock genes. Megakaryocyte-directed deletion of Grin1 in mice caused significant disruption of the circadian rhythm pathway at the transcriptional level and increased expression of BMAL1 at the protein level. Similarly, both pharmacological (MK-801) and genetic (GRIN-/-) inhibition of NMDAR in Meg-01 cells in vitro resulted in widespread changes in clock gene expression including increased expression of BMAL1, the core clock transcription factor. BMAL1 overexpression reduced Meg-01 cell proliferation and altered the time-dependent expression of the cell cycle regulators MYC and WEE1, whereas BMAL1 knockdown led to increased cell death in Meg-01-GRIN1-/- cells. Our results demonstrate that NMDAR regulates the circadian clock in megakaryocytic cells and that the circadian clock component BMAL1 contributes to the control of Meg-01 cell proliferation and survival.
Collapse
Affiliation(s)
- James I Hearn
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Mariam Alhilali
- Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
| | - Minah Kim
- Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
| | - Maggie L Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- Department of Pathology and Laboratory Medicine, Haematology Laboratory, Auckland City Hospital, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
7
|
Draijer S, Timmerman R, Pannekeet J, van Harten A, Farshadi EA, Kemmer J, van Gilst D, Chaves I, Hoekman MFM. FoxO3 Modulates Circadian Rhythms in Neural Stem Cells. Int J Mol Sci 2023; 24:13662. [PMID: 37686468 PMCID: PMC10563086 DOI: 10.3390/ijms241713662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Both FoxO transcription factors and the circadian clock act on the interface of metabolism and cell cycle regulation and are important regulators of cellular stress and stem cell homeostasis. Importantly, FoxO3 preserves the adult neural stem cell population by regulating cell cycle and cellular metabolism and has been shown to regulate circadian rhythms in the liver. However, whether FoxO3 is a regulator of circadian rhythms in neural stem cells remains unknown. Here, we show that loss of FoxO3 disrupts circadian rhythmicity in cultures of neural stem cells, an effect that is mediated via regulation of Clock transcriptional levels. Using Rev-Erbα-VNP as a reporter, we then demonstrate that loss of FoxO3 does not disrupt circadian rhythmicity at the single cell level. A meta-analysis of published data revealed dynamic co-occupancy of multiple circadian clock components within FoxO3 regulatory regions, indicating that FoxO3 is a Clock-controlled gene. Finally, we examined proliferation in the hippocampus of FoxO3-deficient mice and found that loss of FoxO3 delayed the circadian phase of hippocampal proliferation, indicating that FoxO3 regulates correct timing of NSC proliferation. Taken together, our data suggest that FoxO3 is an integral part of circadian regulation of neural stem cell homeostasis.
Collapse
Affiliation(s)
- Swip Draijer
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Raissa Timmerman
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Jesse Pannekeet
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Alexandra van Harten
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Elham Aida Farshadi
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Julius Kemmer
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Demy van Gilst
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Inês Chaves
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Marco F. M. Hoekman
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| |
Collapse
|
8
|
Lathe R, St Clair D. Programmed ageing: decline of stem cell renewal, immunosenescence, and Alzheimer's disease. Biol Rev Camb Philos Soc 2023; 98:1424-1458. [PMID: 37068798 DOI: 10.1111/brv.12959] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
The characteristic maximum lifespan varies enormously across animal species from a few hours to hundreds of years. This argues that maximum lifespan, and the ageing process that itself dictates lifespan, are to a large extent genetically determined. Although controversial, this is supported by firm evidence that semelparous species display evolutionarily programmed ageing in response to reproductive and environmental cues. Parabiosis experiments reveal that ageing is orchestrated systemically through the circulation, accompanied by programmed changes in hormone levels across a lifetime. This implies that, like the circadian and circannual clocks, there is a master 'clock of age' (circavital clock) located in the limbic brain of mammals that modulates systemic changes in growth factor and hormone secretion over the lifespan, as well as systemic alterations in gene expression as revealed by genomic methylation analysis. Studies on accelerated ageing in mice, as well as human longevity genes, converge on evolutionarily conserved fibroblast growth factors (FGFs) and their receptors, including KLOTHO, as well as insulin-like growth factors (IGFs) and steroid hormones, as key players mediating the systemic effects of ageing. Age-related changes in these and multiple other factors are inferred to cause a progressive decline in tissue maintenance through failure of stem cell replenishment. This most severely affects the immune system, which requires constant renewal from bone marrow stem cells. Age-related immune decline increases risk of infection whereas lifespan can be extended in germfree animals. This and other evidence suggests that infection is the major cause of death in higher organisms. Immune decline is also associated with age-related diseases. Taking the example of Alzheimer's disease (AD), we assess the evidence that AD is caused by immunosenescence and infection. The signature protein of AD brain, Aβ, is now known to be an antimicrobial peptide, and Aβ deposits in AD brain may be a response to infection rather than a cause of disease. Because some cognitively normal elderly individuals show extensive neuropathology, we argue that the location of the pathology is crucial - specifically, lesions to limbic brain are likely to accentuate immunosenescence, and could thus underlie a vicious cycle of accelerated immune decline and microbial proliferation that culminates in AD. This general model may extend to other age-related diseases, and we propose a general paradigm of organismal senescence in which declining stem cell proliferation leads to programmed immunosenescence and mortality.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, Chancellor's Building, University of Edinburgh Medical School, Little France, Edinburgh, EH16 4SB, UK
| | - David St Clair
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
9
|
Sato S, Hishida T, Kinouchi K, Hatanaka F, Li Y, Nguyen Q, Chen Y, Wang PH, Kessenbrock K, Li W, Izpisua Belmonte JC, Sassone-Corsi P. The circadian clock CRY1 regulates pluripotent stem cell identity and somatic cell reprogramming. Cell Rep 2023; 42:112590. [PMID: 37261952 DOI: 10.1016/j.celrep.2023.112590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/28/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Distinct metabolic conditions rewire circadian-clock-controlled signaling pathways leading to the de novo construction of signal transduction networks. However, it remains unclear whether metabolic hallmarks unique to pluripotent stem cells (PSCs) are connected to clock functions. Reprogramming somatic cells to a pluripotent state, here we highlighted non-canonical functions of the circadian repressor CRY1 specific to PSCs. Metabolic reprogramming, including AMPK inactivation and SREBP1 activation, was coupled with the accumulation of CRY1 in PSCs. Functional assays verified that CRY1 is required for the maintenance of self-renewal capacity, colony organization, and metabolic signatures. Genome-wide occupancy of CRY1 identified CRY1-regulatory genes enriched in development and differentiation in PSCs, albeit not somatic cells. Last, cells lacking CRY1 exhibit differential gene expression profiles during induced PSC (iPSC) reprogramming, resulting in impaired iPSC reprogramming efficiency. Collectively, these results suggest the functional implication of CRY1 in pluripotent reprogramming and ontogenesis, thereby dictating PSC identity.
Collapse
Affiliation(s)
- Shogo Sato
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA; Center for Biological Clocks Research, Department of Biology, Texas A&M University, College Station, TX, USA.
| | - Tomoaki Hishida
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA; Laboratory of Biological Chemistry, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Kenichiro Kinouchi
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Fumiaki Hatanaka
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA; Altos Labs, San Diego, CA, USA
| | - Yumei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Quy Nguyen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Yumay Chen
- UC Irvine Diabetes Center, Sue and Bill Gross Stem Cell Research Center, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ping H Wang
- UC Irvine Diabetes Center, Sue and Bill Gross Stem Cell Research Center, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA; Altos Labs, San Diego, CA, USA.
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
10
|
Chopra K, Folkmanaitė M, Stockdale L, Shathish V, Ishibashi S, Bergin R, Amich J, Amaya E. Duox is the primary NADPH oxidase responsible for ROS production during adult caudal fin regeneration in zebrafish. iScience 2023; 26:106147. [PMID: 36843843 PMCID: PMC9950526 DOI: 10.1016/j.isci.2023.106147] [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: 04/26/2022] [Revised: 11/28/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Sustained elevated levels of reactive oxygen species (ROS) have been shown to be essential for regeneration in many organisms. This has been shown primarily via the use of pharmacological inhibitors targeting the family of NADPH oxidases (NOXes). To identify the specific NOXes involved in ROS production during adult caudal fin regeneration in zebrafish, we generated nox mutants for duox, nox5 and cyba (a key subunit of NOXes 1-4) and crossed these lines with a transgenic line ubiquitously expressing HyPer, which permits the measurement of ROS levels. Homozygous duox mutants had the greatest effect on ROS levels and rate of fin regeneration among the single mutants. However, duox:cyba double mutants showed a greater effect on fin regeneration than the single duox mutants, suggesting that Nox1-4 also play a role during regeneration. This work also serendipitously found that ROS levels in amputated adult zebrafish fins oscillate with a circadian rhythm.
Collapse
Affiliation(s)
- Kunal Chopra
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Milda Folkmanaitė
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Liam Stockdale
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Vishali Shathish
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Shoko Ishibashi
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Rachel Bergin
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Jorge Amich
- Manchester Fungal Infection Group (MFIG), Division of Evolution, Infection, and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK.,Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Majadahonda 28220 Madrid, Spain
| | - Enrique Amaya
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
11
|
Ji S, Xiong M, Chen H, Liu Y, Zhou L, Hong Y, Wang M, Wang C, Fu X, Sun X. Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:116. [PMID: 36918530 PMCID: PMC10015098 DOI: 10.1038/s41392-023-01343-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 01/19/2023] [Indexed: 03/16/2023] Open
Abstract
The ageing process is a systemic decline from cellular dysfunction to organ degeneration, with more predisposition to deteriorated disorders. Rejuvenation refers to giving aged cells or organisms more youthful characteristics through various techniques, such as cellular reprogramming and epigenetic regulation. The great leaps in cellular rejuvenation prove that ageing is not a one-way street, and many rejuvenative interventions have emerged to delay and even reverse the ageing process. Defining the mechanism by which roadblocks and signaling inputs influence complex ageing programs is essential for understanding and developing rejuvenative strategies. Here, we discuss the intrinsic and extrinsic factors that counteract cell rejuvenation, and the targeted cells and core mechanisms involved in this process. Then, we critically summarize the latest advances in state-of-art strategies of cellular rejuvenation. Various rejuvenation methods also provide insights for treating specific ageing-related diseases, including cellular reprogramming, the removal of senescence cells (SCs) and suppression of senescence-associated secretory phenotype (SASP), metabolic manipulation, stem cells-associated therapy, dietary restriction, immune rejuvenation and heterochronic transplantation, etc. The potential applications of rejuvenation therapy also extend to cancer treatment. Finally, we analyze in detail the therapeutic opportunities and challenges of rejuvenation technology. Deciphering rejuvenation interventions will provide further insights into anti-ageing and ageing-related disease treatment in clinical settings.
Collapse
Affiliation(s)
- Shuaifei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Huating Chen
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiqiong Liu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Laixian Zhou
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China.
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| |
Collapse
|
12
|
Bahiru MS, Bittman EL. Adult Neurogenesis Is Altered by Circadian Phase Shifts and the Duper Mutation in Female Syrian Hamsters. eNeuro 2023; 10:ENEURO.0359-22.2023. [PMID: 36878716 PMCID: PMC10062491 DOI: 10.1523/eneuro.0359-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Cell birth and survival in the adult hippocampus are regulated by a circadian clock. Rotating shift work and jet lag disrupt circadian rhythms and aggravate disease. Internal misalignment, a state in which abnormal phase relationships prevail between and within organs, is proposed to account for adverse effects of circadian disruption. This hypothesis has been difficult to test because phase shifts of the entraining cycle inevitably lead to transient desynchrony. Thus, it remains possible that phase shifts, regardless of internal desynchrony, account for adverse effects of circadian disruption and alter neurogenesis and cell fate. To address this question, we examined cell birth and differentiation in the duper Syrian hamster (Mesocricetus auratus), a Cry1-null mutant in which re-entrainment of locomotor rhythms is greatly accelerated. Adult females were subjected to alternating 8 h advances and delays at eight 16 d intervals. BrdU, a cell birth marker, was given midway through the experiment. Repeated phase shifts decreased the number of newborn non-neuronal cells in WT, but not in duper hamsters. The duper mutation increased the number of BrdU-IR cells that stained for NeuN, which marks neuronal differentiation. Immunocytochemical staining for proliferating cell nuclear antigen indicated no overall effect of genotype or repeated shifts on cell division rates after 131 days. Cell differentiation, assessed by doublecortin, was higher in duper hamsters but was not significantly altered by repeated phase shifts. Our results support the internal misalignment hypothesis and indicate that Cry1 regulates cell differentiation. Phase shifts may determine neuronal stem cell survival and time course of differentiation after cell birth. Figure created with BioRender.
Collapse
Affiliation(s)
- Michael Seifu Bahiru
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts 01003
| | - Eric L Bittman
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts 01003
- Department of Biology, University of Massachusetts, Amherst, Massachusetts 01003
| |
Collapse
|
13
|
Clements A, Shibuya Y, Hokugo A, Brooks Z, Roca Y, Kondo T, Nishimura I, Jarrahy R. In vitro assessment of Neuronal PAS domain 2 mitigating compounds for scarless wound healing. Front Med (Lausanne) 2023; 9:1014763. [PMID: 36816724 PMCID: PMC9928850 DOI: 10.3389/fmed.2022.1014763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/09/2022] [Indexed: 02/04/2023] Open
Abstract
Background The core circadian gene Neuronal PAS domain 2 (NPAS2) is expressed in dermal fibroblasts and has been shown to play a critical role in regulating collagen synthesis during wound healing. We have performed high throughput drug screening to identify genes responsible for downregulation of Npas2 while maintaining cell viability. From this, five FDA-approved hit compounds were shown to suppress Npas2 expression in fibroblasts. In this study, we hypothesize that the therapeutic suppression of Npas2 by hit compounds will have two effects: (1) attenuated excessive collagen deposition and (2) accelerated dermal wound healing without hypertrophic scarring. Materials and methods To test the effects of each hit compound (named Dwn1, 2, 3, 4, and 5), primary adult human dermal fibroblasts (HDFa) were treated with either 0, 0.1, 1, or 10 μM of a single hit compound. HDFa behaviors were assessed by picrosirius red staining and quantitative RT-PCR for in vitro collagen synthesis, cell viability assay, in vitro fibroblast-to-myofibroblast differentiation test, and cell migration assays. Results Dwn1 and Dwn2 were found to significantly affect collagen synthesis and cell migration without any cytotoxicity. Dwn3, Dwn4, and Dwn5 did not affect collagen synthesis and were thereby eliminated from further consideration for their role in mitigation of gene expression or myofibroblast differentiation. Dwn1 also attenuated myofibroblast differentiation on HDFa. Conclusion Dwn1 and Dwn2 may serve as possible therapeutic agents for future studies related to skin wound healing.
Collapse
Affiliation(s)
- Adam Clements
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yoichiro Shibuya
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Akishige Hokugo
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States,*Correspondence: Akishige Hokugo,
| | - Zachary Brooks
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yvonne Roca
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Takeru Kondo
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ichiro Nishimura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States,Ichiro Nishimura,
| | - Reza Jarrahy
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States,Reza Jarrahy,
| |
Collapse
|
14
|
Vanoni EM, Nandrot EF. The Retinal Pigment Epithelium: Cells That Know the Beat! ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:539-545. [PMID: 37440084 DOI: 10.1007/978-3-031-27681-1_79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The retinal pigment epithelium (RPE) ensures different functions crucial for photoreceptor survival, and thus for vision, such as photoreceptor outer segments (POS) phagocytosis and retinal adhesion. Both follow a circadian rhythm with an activity peak occurring respectively 1.5-2 and 3.5 h after light onset. Interestingly, we showed that two rodent models, β5-/- and Prpf31+/- mice, display distinct alterations in both functions leading to different phenotypes. Indeed, the phagocytic peak totally disappears in β5 knockout mice but is attenuated and shifted in Prpf31+/- mice. Conversely, the retinal adhesion peak only attenuated in β5-/- mice is lost in Prpf31+/- mice. These distinct alterations have different consequences on retinal homeostasis proportional to the observed defects: β5-/- mice progressively lose vision and accumulate RPE lipofuscin deposits, while Prpf31+/- mice develop RPE metabolic dysfunctions and gradual structural modifications indicative of cellular stress. Hence, animal models are useful to understand the importance of the proper regulation of these functions.
Collapse
Affiliation(s)
- Elora M Vanoni
- Therapeutics Department, Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Emeline F Nandrot
- Therapeutics Department, Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.
| |
Collapse
|
15
|
Rowton M, Perez-Cervantes C, Hur S, Jacobs-Li J, Lu E, Deng N, Guzzetta A, Hoffmann AD, Stocker M, Steimle JD, Lazarevic S, Oubaha S, Yang XH, Kim C, Yu S, Eckart H, Koska M, Hanson E, Chan SSK, Garry DJ, Kyba M, Basu A, Ikegami K, Pott S, Moskowitz IP. Hedgehog signaling activates a mammalian heterochronic gene regulatory network controlling differentiation timing across lineages. Dev Cell 2022; 57:2181-2203.e9. [PMID: 36108627 PMCID: PMC10506397 DOI: 10.1016/j.devcel.2022.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/24/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022]
Abstract
Many developmental signaling pathways have been implicated in lineage-specific differentiation; however, mechanisms that explicitly control differentiation timing remain poorly defined in mammals. We report that murine Hedgehog signaling is a heterochronic pathway that determines the timing of progenitor differentiation. Hedgehog activity was necessary to prevent premature differentiation of second heart field (SHF) cardiac progenitors in mouse embryos, and the Hedgehog transcription factor GLI1 was sufficient to delay differentiation of cardiac progenitors in vitro. GLI1 directly activated a de novo progenitor-specific network in vitro, akin to that of SHF progenitors in vivo, which prevented the onset of the cardiac differentiation program. A Hedgehog signaling-dependent active-to-repressive GLI transition functioned as a differentiation timer, restricting the progenitor network to the SHF. GLI1 expression was associated with progenitor status across germ layers, and it delayed the differentiation of neural progenitors in vitro, suggesting a broad role for Hedgehog signaling as a heterochronic pathway.
Collapse
Affiliation(s)
- Megan Rowton
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Carlos Perez-Cervantes
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Suzy Hur
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Jessica Jacobs-Li
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Emery Lu
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Nikita Deng
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Alexander Guzzetta
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Andrew D Hoffmann
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Matthew Stocker
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Jeffrey D Steimle
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Sonja Lazarevic
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Sophie Oubaha
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Xinan H Yang
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Chul Kim
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Shuhan Yu
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Heather Eckart
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Mervenaz Koska
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Erika Hanson
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Sunny S K Chan
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel J Garry
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anindita Basu
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Kohta Ikegami
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Sebastian Pott
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA
| | - Ivan P Moskowitz
- Departments of Pediatrics, Pathology, Human Genetics, and Genetic Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Hart DA, Zernicke RF, Shrive NG. Homo sapiens May Incorporate Daily Acute Cycles of “Conditioning–Deconditioning” to Maintain Musculoskeletal Integrity: Need to Integrate with Biological Clocks and Circadian Rhythm Mediators. Int J Mol Sci 2022; 23:ijms23179949. [PMID: 36077345 PMCID: PMC9456265 DOI: 10.3390/ijms23179949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022] Open
Abstract
Human evolution required adaptation to the boundary conditions of Earth, including 1 g gravity. The bipedal mobility of Homo sapiens in that gravitational field causes ground reaction force (GRF) loading of their lower extremities, influencing the integrity of the tissues of those extremities. However, humans usually experience such loading during the day and then a period of relative unloading at night. Many studies have indicated that loading of tissues and cells of the musculoskeletal (MSK) system can inhibit their responses to biological mediators such as cytokines and growth factors. Such findings raise the possibility that humans use such cycles of acute conditioning and deconditioning of the cells and tissues of the MSK system to elaborate critical mediators and responsiveness in parallel with these cycles, particularly involving GRF loading. However, humans also experience circadian rhythms with the levels of a number of mediators influenced by day/night cycles, as well as various levels of biological clocks. Thus, if responsiveness to MSK-generated mediators also occurs during the unloaded part of the daily cycle, that response must be integrated with circadian variations as well. Furthermore, it is also possible that responsiveness to circadian rhythm mediators may be regulated by MSK tissue loading. This review will examine evidence for the above scenario and postulate how interactions could be both regulated and studied, and how extension of the acute cycles biased towards deconditioning could lead to loss of tissue integrity.
Collapse
Affiliation(s)
- David A. Hart
- Department of Surgery, University of Calgary, Calgary, AB T2N 4N1, Canada
- McCaig Institute for Bone & Joint Health Research, University of Calgary, Calgary, AB T2N 4N1, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Bone & Joint Health Strategic Clinical Network, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
- Correspondence:
| | - Ronald F. Zernicke
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI 48109-5328, USA
- School of Kinesiology, University of Michigan, Ann Arbor, MI 48108-1048, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109-2099, USA
| | - Nigel G. Shrive
- Department of Surgery, University of Calgary, Calgary, AB T2N 4N1, Canada
- McCaig Institute for Bone & Joint Health Research, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Civil Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB T2N 4V8, Canada
| |
Collapse
|
17
|
Poljšak B, Kovač V, Milisav I. Current Uncertainties and Future Challenges Regarding NAD+ Boosting Strategies. Antioxidants (Basel) 2022; 11:1637. [PMID: 36139711 PMCID: PMC9495723 DOI: 10.3390/antiox11091637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Precursors of nicotinamide adenine dinucleotide (NAD+), modulators of enzymes of the NAD+ biosynthesis pathways and inhibitors of NAD+ consuming enzymes, are the main boosters of NAD+. Increasing public awareness and interest in anti-ageing strategies and health-promoting lifestyles have grown the interest in the use of NAD+ boosters as dietary supplements, both in scientific circles and among the general population. Here, we discuss the current trends in NAD+ precursor usage as well as the uncertainties in dosage, timing, safety, and side effects. There are many unknowns regarding pharmacokinetics and pharmacodynamics, particularly bioavailability, metabolism, and tissue specificity of NAD+ boosters. Given the lack of long-term safety studies, there is a need for more clinical trials to determine the proper dose of NAD+ boosters and treatment duration for aging prevention and as disease therapy. Further research will also need to address the long-term consequences of increased NAD+ and the best approaches and combinations to increase NAD+ levels. The answers to the above questions will contribute to the more efficient and safer use of NAD+ boosters.
Collapse
Affiliation(s)
- Borut Poljšak
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Vito Kovač
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Irina Milisav
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
18
|
A guiding role of the Arabidopsis circadian clock in cell differentiation revealed by time-series single-cell RNA sequencing. Cell Rep 2022; 40:111059. [PMID: 35830805 DOI: 10.1016/j.celrep.2022.111059] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 04/01/2022] [Accepted: 06/14/2022] [Indexed: 11/21/2022] Open
Abstract
Circadian rhythms and progression of cell differentiation are closely coupled in multicellular organisms. However, whether establishment of circadian rhythms regulates cell differentiation or vice versa has not been elucidated due to technical limitations. Here, we exploit high cell fate plasticity of plant cells to perform single-cell RNA sequencing during the entire process of cell differentiation. By analyzing reconstructed actual time series of the differentiation processes at single-cell resolution using a method we developed (PeakMatch), we find that the expression profile of clock genes is changed prior to cell differentiation, including induction of the clock gene LUX ARRYTHMO (LUX). ChIP sequencing analysis reveals that LUX induction in early differentiating cells directly targets genes involved in cell-cycle progression to regulate cell differentiation. Taken together, these results not only reveal a guiding role of the plant circadian clock in cell differentiation but also provide an approach for time-series analysis at single-cell resolution.
Collapse
|
19
|
Gao W, Li R, Ye M, Zhang L, Zheng J, Yang Y, Wei X, Zhao Q. The circadian clock has roles in mesenchymal stem cell fate decision. Stem Cell Res Ther 2022; 13:200. [PMID: 35578353 PMCID: PMC9109355 DOI: 10.1186/s13287-022-02878-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/26/2022] [Indexed: 02/08/2023] Open
Abstract
The circadian clock refers to the intrinsic biological rhythms of physiological functions and behaviours. It synergises with the solar cycle and has profound effects on normal metabolism and organismal fitness. Recent studies have suggested that the circadian clock exerts great influence on the differentiation of stem cells. Here, we focus on the close relationship between the circadian clock and mesenchymal stem cell fate decisions in the skeletal system. The underlying mechanisms include hormone signals and the activation and repression of different transcription factors under circadian regulation. Additionally, the clock interacts with epigenetic modifiers and non-coding RNAs and is even involved in chromatin remodelling. Although the specificity and safety of circadian therapy need to be further studied, the circadian regulation of stem cells can be regarded as a promising candidate for health improvement and disease prevention.
Collapse
Affiliation(s)
- Wenzhen Gao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Rong Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Meilin Ye
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, School and Hospital of Stomatology, Shandong University, Jinan, 250012, China
| | - Lanxin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiawen Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuqing Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoyu Wei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qing Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
20
|
Bery A, Bagchi U, Bergen AA, Felder-Schmittbuhl MP. Circadian clocks, retinogenesis and ocular health in vertebrates: new molecular insights. Dev Biol 2022; 484:40-56. [DOI: 10.1016/j.ydbio.2022.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 12/22/2022]
|
21
|
Ghanemi A, Yoshioka M, St-Amand J. Exercise, Diet and Sleeping as Regenerative Medicine Adjuvants: Obesity and Ageing as Illustrations. MEDICINES (BASEL, SWITZERLAND) 2022; 9:medicines9010007. [PMID: 35049940 PMCID: PMC8778846 DOI: 10.3390/medicines9010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/21/2022]
Abstract
Regenerative medicine uses the biological and medical knowledge on how the cells and tissue regenerate and evolve in order to develop novel therapies. Health conditions such as ageing, obesity and cancer lead to an impaired regeneration ability. Exercise, diet choices and sleeping pattern have significant impacts on regeneration biology via diverse pathways including reducing the inflammatory and oxidative components. Thus, exercise, diet and sleeping management can be optimized towards therapeutic applications in regenerative medicine. It could allow to prevent degeneration, optimize the biological regeneration and also provide adjuvants for regenerative medicine.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
| | - Jonny St-Amand
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-654-2296
| |
Collapse
|
22
|
Kahn M. Taking the road less traveled - the therapeutic potential of CBP/β-catenin antagonists. Expert Opin Ther Targets 2021; 25:701-719. [PMID: 34633266 PMCID: PMC8745629 DOI: 10.1080/14728222.2021.1992386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 10/20/2022]
Abstract
AREAS COVERED This perspective discusses the challenges of targeting the Wnt signaling cascade, the safety, efficacy, and therapeutic potential of specific CBP/β-catenin antagonists and a rationale for the pleiotropic effects of CBP/β-catenin antagonists beyond Wnt signaling. EXPERT OPINION CBP/β-catenin antagonists can correct lineage infidelity, enhance wound healing, both normal and aberrant (e.g. fibrosis) and force the differentiation and lineage commitment of stem cells and cancer stem cells by regulating enhancer and super-enhancer coactivator occupancy. Small molecule CBP/β-catenin antagonists rebalance the equilibrium between CBP/β-catenin versus p300/β-catenin dependent transcription and may be able to treat or prevent many diseases of aging, via maintenance of our somatic stem cell pool, and regulating mitochondrial function and metabolism involved in differentiation and immune cell function.
Collapse
Affiliation(s)
- Michael Kahn
- Department of Molecular Medicine, City of Hope, Beckman Research Institute, 1500 East Duarte Road Flower Building, Duarte, CA, USA
| |
Collapse
|
23
|
Poulsen RC, Hearn JI, Dalbeth N. The circadian clock: a central mediator of cartilage maintenance and osteoarthritis development? Rheumatology (Oxford) 2021; 60:3048-3057. [PMID: 33630038 DOI: 10.1093/rheumatology/keab197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/20/2021] [Indexed: 01/03/2023] Open
Abstract
The circadian clock is a specialized cell signalling pathway present in all cells. Loss of clock function leads to tissue degeneration and premature ageing in animal models demonstrating the fundamental importance of clocks for cell, tissue and organism health. There is now considerable evidence that the chondrocyte circadian clock is altered in OA. The purpose of this review is to summarize current knowledge regarding the nature of the change in the chondrocyte clock in OA and the implications of this change for disease development. Expression of the core clock component, BMAL1, has consistently been shown to be lower in OA chondrocytes. This may contribute to changes in chondrocyte differentiation and extracellular matrix turnover in disease. Circadian clocks are highly responsive to environmental factors. Mechanical loading, diet, inflammation and oxidative insult can all influence clock function. These factors may contribute to causing the change in the chondrocyte clock in OA.
Collapse
Affiliation(s)
- Raewyn C Poulsen
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences.,Department of Medicine, School of Medicine
| | - James I Hearn
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
24
|
Garikipati VNS, Arakelyan A, Blakely EA, Chang PY, Truongcao MM, Cimini M, Malaredy V, Bajpai A, Addya S, Bisserier M, Brojakowska A, Eskandari A, Khlgatian MK, Hadri L, Fish KM, Kishore R, Goukassian DA. Long-Term Effects of Very Low Dose Particle Radiation on Gene Expression in the Heart: Degenerative Disease Risks. Cells 2021; 10:387. [PMID: 33668521 PMCID: PMC7917872 DOI: 10.3390/cells10020387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/27/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
Compared to low doses of gamma irradiation (γ-IR), high-charge-and-energy (HZE) particle IR may have different biological response thresholds in cardiac tissue at lower doses, and these effects may be IR type and dose dependent. Three- to four-month-old female CB6F1/Hsd mice were exposed once to one of four different doses of the following types of radiation: γ-IR 137Cs (40-160 cGy, 0.662 MeV), 14Si-IR (4-32 cGy, 260 MeV/n), or 22Ti-IR (3-26 cGy, 1 GeV/n). At 16 months post-exposure, animals were sacrificed and hearts were harvested and archived as part of the NASA Space Radiation Tissue Sharing Forum. These heart tissue samples were used in our study for RNA isolation and microarray hybridization. Functional annotation of twofold up/down differentially expressed genes (DEGs) and bioinformatics analyses revealed the following: (i) there were no clear lower IR thresholds for HZE- or γ-IR; (ii) there were 12 common DEGs across all 3 IR types; (iii) these 12 overlapping genes predicted various degrees of cardiovascular, pulmonary, and metabolic diseases, cancer, and aging; and (iv) these 12 genes revealed an exclusive non-linear DEG pattern in 14Si- and 22Ti-IR-exposed hearts, whereas two-thirds of γ-IR-exposed hearts revealed a linear pattern of DEGs. Thus, our study may provide experimental evidence of excess relative risk (ERR) quantification of low/very low doses of full-body space-type IR-associated degenerative disease development.
Collapse
Affiliation(s)
- Venkata Naga Srikanth Garikipati
- Department of Emergency Medicine, Dorothy M Davis Heart and Lung Research Institute, Wexner Medical School, The Ohio State University, Columbus, OH 43210, USA;
| | - Arsen Arakelyan
- Bioinformatics Group, The Institute of Molecular Biology, The National Academy of Sciences of the Republic of Armenia, Yerevan 0014, Armenia;
- PathVerse, Yerevan 0014, Armenia
| | | | | | - May M. Truongcao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Maria Cimini
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Vandana Malaredy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Anamika Bajpai
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - Sankar Addya
- Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Agnieszka Brojakowska
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Abrisham Eskandari
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Mary K. Khlgatian
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Kenneth M. Fish
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.M.T.); (M.C.); (V.M.); (A.B.); (R.K.)
| | - David. A. Goukassian
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.B.); (A.B.); (A.E.); (M.K.K.); (L.H.); (K.M.F.)
| |
Collapse
|
25
|
Litovchenko M, Meireles-Filho ACA, Frochaux MV, Bevers RPJ, Prunotto A, Anduaga AM, Hollis B, Gardeux V, Braman VS, Russeil JMC, Kadener S, Dal Peraro M, Deplancke B. Extensive tissue-specific expression variation and novel regulators underlying circadian behavior. SCIENCE ADVANCES 2021; 7:eabc3781. [PMID: 33514540 PMCID: PMC7846174 DOI: 10.1126/sciadv.abc3781] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 12/10/2020] [Indexed: 05/10/2023]
Abstract
Natural genetic variation affects circadian rhythms across the evolutionary tree, but the underlying molecular mechanisms are poorly understood. We investigated population-level, molecular circadian clock variation by generating >700 tissue-specific transcriptomes of Drosophila melanogaster (w1118 ) and 141 Drosophila Genetic Reference Panel (DGRP) lines. This comprehensive circadian gene expression atlas contains >1700 cycling genes including previously unknown central circadian clock components and tissue-specific regulators. Furthermore, >30% of DGRP lines exhibited aberrant circadian gene expression, revealing abundant genetic variation-mediated, intertissue circadian expression desynchrony. Genetic analysis of one line with the strongest deviating circadian expression uncovered a novel cry mutation that, as shown by protein structural modeling and brain immunohistochemistry, disrupts the light-driven flavin adenine dinucleotide cofactor photoreduction, providing in vivo support for the importance of this conserved photoentrainment mechanism. Together, our study revealed pervasive tissue-specific circadian expression variation with genetic variants acting upon tissue-specific regulatory networks to generate local gene expression oscillations.
Collapse
Affiliation(s)
- Maria Litovchenko
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Antonio C A Meireles-Filho
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Michael V Frochaux
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Roel P J Bevers
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Alessio Prunotto
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | | | - Brian Hollis
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Vincent Gardeux
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Virginie S Braman
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Julie M C Russeil
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | | | - Matteo Dal Peraro
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| | - Bart Deplancke
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Vaud 1015, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Vaud, Switzerland
| |
Collapse
|
26
|
Huang S, Choi MH, Huang H, Wang X, Chang YC, Kim JY. Demyelination Regulates the Circadian Transcription Factor BMAL1 to Signal Adult Neural Stem Cells to Initiate Oligodendrogenesis. Cell Rep 2020; 33:108394. [PMID: 33207207 DOI: 10.1016/j.celrep.2020.108394] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/10/2020] [Accepted: 10/26/2020] [Indexed: 10/22/2022] Open
Abstract
Circadian clocks are endogenous oscillators that generate cell-autonomous rhythms that govern cellular processes and are synchronized by external cues in the local macro- and micro-environments. Demyelination, a common brain pathology with variable degrees of recovery, changes the microenvironment via damaged myelin and activation of glial cells. How these microenvironmental changes affect local circadian clocks and with what consequences is mostly unknown. Here, we show that within demyelinating lesions, astrocyte circadian clocks produce the Wnt inhibitors SFRP1 and SFRP5. Unexpectedly, SFRP1 and SFRP5 signal to the subventricular zone (SVZ) to reduce the circadian transcription factor BMAL1. This sequence of events causes adult neural stem cells in the SVZ to differentiate into oligodendrocyte lineage cells, which are then supplied to demyelinated lesions. Our findings show that circadian clocks in demyelinating lesions respond to microenvironmental changes and communicate with the SVZ to enhance a natural repair system of spontaneous remyelination.
Collapse
Affiliation(s)
- Suihong Huang
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China
| | - Ming Ho Choi
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Hao Huang
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Yu Chen Chang
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China
| | - Jin Young Kim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
27
|
Malaguarnera R, Ledda C, Filippello A, Frasca F, Francavilla VC, Ramaci T, Parisi MC, Rapisarda V, Piro S. Thyroid Cancer and Circadian Clock Disruption. Cancers (Basel) 2020; 12:E3109. [PMID: 33114365 PMCID: PMC7690860 DOI: 10.3390/cancers12113109] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Thyroid cancer (TC) represents the most common malignancy of the endocrine system, with an increased incidence across continents attributable to both improvement of diagnostic procedures and environmental factors. Among the modifiable risk factors, insulin resistance might influence the development of TC. A relationship between circadian clock machinery disfunction and TC has recently been proposed. The circadian clock machinery comprises a set of rhythmically expressed genes responsible for circadian rhythms. Perturbation of this system contributes to the development of pathological states such as cancer. Several clock genes have been found deregulated upon thyroid nodule malignant transformation. The molecular mechanisms linking circadian clock disruption and TC are still unknown but could include insulin resistance. Circadian misalignment occurring during shift work, jet lag, high fat food intake, is associated with increased insulin resistance. This metabolic alteration, in turn, is associated with a well-known risk factor for TC i.e., hyperthyrotropinemia, which could also be induced by sleep disturbances. In this review, we describe the mechanisms controlling the circadian clock function and its involvement in the cell cycle, stemness and cancer. Moreover, we discuss the evidence supporting the link between circadian clockwork disruption and TC development/progression, highlighting its potential implications for TC prevention, diagnosis and therapy.
Collapse
Affiliation(s)
- Roberta Malaguarnera
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Caterina Ledda
- Department of Clinical and Experimental Medicine, Occupational Medicine, University of Catania, 95100 Catania, Italy;
| | - Agnese Filippello
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (A.F.); (S.P.)
| | - Francesco Frasca
- Endocrinology Unit, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy;
| | - Vincenzo Cristian Francavilla
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Tiziana Ramaci
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Maria Chiara Parisi
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Venerando Rapisarda
- Department of Clinical and Experimental Medicine, Occupational Medicine, University of Catania, 95100 Catania, Italy;
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (A.F.); (S.P.)
| |
Collapse
|
28
|
Effect of Type 2 Diabetes Mellitus on the Hypoxia-Inducible Factor 1-Alpha Expression. Is There a Relationship with the Clock Genes? J Clin Med 2020; 9:jcm9082632. [PMID: 32823749 PMCID: PMC7465909 DOI: 10.3390/jcm9082632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 01/21/2023] Open
Abstract
Limited reports exist on the relationships between regulation of oxygen homeostasis and circadian clock genes in type 2 diabetes. We examined whether the expression of Hypoxia-Inducible Factor-1α (HIF-1α) and HIF-2α relates to changes in the expression of clock genes (Period homolog proteins (PER)1, PER2, PER3, Retinoid-related orphan receptor alpha (RORA), Aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL), Circadian locomotor output cycles kaput (CLOCK), and Cryptochrome proteins (CRY) 1 and CRY2) in patients with type 2 diabetes. A total of 129 subjects were evaluated in this cross-sectional study (48% with diabetes). The gene expression was measured by polymerase chain reaction. The lactate and pyruvate levels were used as surrogate of the hypoxia induced anaerobic glycolysis activity. Patients with diabetes showed an increased plasma concentration of both lactate (2102.1 ± 688.2 vs. 1730.4 ± 694.4 uM/L, p = 0.013) and pyruvate (61.9 ± 25.6 vs. 50.3 ± 23.1 uM/L, p = 0.026) in comparison to controls. However, this finding was accompanied by a blunted HIF-1α expression (1.1 (0.2 to 5.0) vs. 1.7 (0.4 to 9.2) arbitrary units (AU), p ≤ 0.001). Patients with diabetes also showed a significant reduction of all assessed clock genes’ expression. Univariate analysis showed that HIF-1α and almost all clock genes were significantly and negatively correlated with HbA1c concentration. In addition, positive correlations between HIF-1α and the clock genes were observed. The stepwise multivariate regression analysis showed that HbA1c and clock genes independently predicted the expression of HIF-1α. Type 2 diabetes modifies the expression of HIF-1α and clock genes, which correlates with the degree of metabolic control.
Collapse
|
29
|
García-García A, Méndez-Ferrer S. The Autonomic Nervous System Pulls the Strings to Coordinate Circadian HSC Functions. Front Immunol 2020; 11:956. [PMID: 32508835 PMCID: PMC7251159 DOI: 10.3389/fimmu.2020.00956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/23/2020] [Indexed: 11/16/2022] Open
Abstract
As for many other adult stem cells, the behavior of hematopoietic stem and progenitor cells (HSPCs) is subjected to circadian regulatory patterns. Multiple HSPC functions, such as proliferation, differentiation or trafficking exhibit time-dependent patterns that require a tight coordination to ensure daily blood cell production. The autonomic nervous system, together with circulating hormones, relay circadian signals from the central clock-the suprachiasmatic nucleus in the brain-to synchronize HSC niche physiology according to light/darkness cycles. Research over the last 20 years has revealed how specific neural signals modulate certain aspects of circadian HSC biology. However, only recently some studies have started to decipher the cellular and molecular mechanisms that orchestrate this complex regulation in a time-dependent fashion. Here we firstly review some of the recent key findings illustrating how different neural signals (catecholaminergic or cholinergic) regulate circadian HSC egress, homing, maintenance, proliferation, and differentiation. In particular, we highlight the critical role of different neurotransmitter receptors in the bone marrow microenvironment to channel these neural signals and regulate antagonistic processes according to circadian cues and organismal demands. Then, we discuss the potential biological meaning of HSC circadian regulation and its possible utility for clinical purposes. Finally, we offer our perspective on emerging concepts in HSC chronobiology.
Collapse
Affiliation(s)
- Andrés García-García
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Simón Méndez-Ferrer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
30
|
Thakur S, Storewala P, Basak U, Jalan N, Pethe P. Clocking the circadian genes in human embryonic stem cells. Stem Cell Investig 2020; 7:9. [PMID: 32695802 PMCID: PMC7367470 DOI: 10.21037/sci-2020-014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 04/30/2020] [Indexed: 02/05/2023]
Abstract
Multicellular organisms respond to changing environment which is primarily driven by light from the sun. Essential cyclical processes such as digestion, sleep, migration and breeding are controlled by set of genes know as circadian genes. The core circadian genes comprise of CLOCK, BMAL-1, PERIOD and CYRPTOCHROME that are expressed cyclically and they regulate expression of several genes downstream. The expression of circadian genes has been well studied in multicellular animals; however, it has been shown that stem cells also possess active circadian cycle genes. The circadian cycle genes have been studied in mouse embryonic stem cells and in adult human stem cells. However, there are only few reports of circadian cycle genes in human pluripotent stem cells. We used human embryonic stem cells to investigate the expression of CLOCK, BMAL-1, PERIOD and CYRPTOCHORME genes by RT-PCR at 6, 18 and 22 hours in undifferentiated and differentiated cells. We differentiated human embryonic stem cells spontaneously by adding 10% fetal bovine serum (FBS), and the cells primarily differentiated into ectoderm and mesoderm. We report that CLOCK and BMAL-1 are differentially expressed while PERIOD and CRYPTOCHROME show cyclicity in differentiated and undifferentiated cells. Our results show circadian genes are active in human embryonic stem cells and this needs to be further investigated as human pluripotent stem cells have potential to be used for cell therapy, where they need to synchronize with the body's circadian cycle.
Collapse
Affiliation(s)
- Soumyaa Thakur
- NMIMS Sunandan Divatia School of Science, NMIMS (deemed to-be) University, Mumbai, India
| | - Prachi Storewala
- NMIMS Sunandan Divatia School of Science, NMIMS (deemed to-be) University, Mumbai, India
| | - Upasna Basak
- NMIMS Sunandan Divatia School of Science, NMIMS (deemed to-be) University, Mumbai, India
| | - Nitya Jalan
- NMIMS School of Business Management, NMIMS (deemed to-be) University, Mumbai, India
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International University, Pune, India
| |
Collapse
|
31
|
The effect of artificial light at night on the biomass of caterpillars feeding in urban tree canopies. Urban Ecosyst 2020. [DOI: 10.1007/s11252-020-00999-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractAlternation of day and night is the oldest cycle on Earth, which is increasingly disturbed by the accelerating rate of urbanization and technological development. Despite the ubiquity of light pollution in cities, many aspects of its influence on urban ecosystems are still poorly understood. Here we studied the effect of artificial light at night (ALAN) on the biomass of arboreal caterpillar populations, which are a major component of the diet of many insectivorous animals. We predicted that increasing ALAN intensity is associated with reduced caterpillar biomass, because ALAN may increase predation risk for both caterpillars and adult lepidopterans (i.e. moths), and can also hinder the moths’ reproductive rate. We estimated caterpillar biomass from frass samples (n = 3061) collected from 36 focal trees in two cities in Hungary during four consecutive years. To quantify ALAN we measured light intensity during night at each focal tree (range of illumination: 0.69–3.18 lx). We found that caterpillar biomass of individual trees was repeatable over the four years. This temporal consistency in prey biomass production may be important for birds because it can help predict territory quality, especially in cities where caterpillar abundance is generally low. Our results did not support the negative effect of ALAN on urban caterpillar populations, because ALAN intensity was not related to caterpillar biomass, and this lack of effect was consistent between study sites and tree species. We suggest that the effect of ALAN on urban caterpillar biomass is either weak and thus can be masked by other, local environmental factors, or light pollution may have antagonistic effects acting during different stages of the lepidopteran life cycle. Another explanation could be that even the lower levels of our sites’ public lighting are strong enough to cause serious detrimental effects for caterpillars, resulting in their uniformly low biomass.
Collapse
|
32
|
Parasram K, Karpowicz P. Time after time: circadian clock regulation of intestinal stem cells. Cell Mol Life Sci 2020; 77:1267-1288. [PMID: 31586240 PMCID: PMC11105114 DOI: 10.1007/s00018-019-03323-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/16/2019] [Accepted: 09/25/2019] [Indexed: 12/22/2022]
Abstract
Daily fluctuations in animal physiology, known as circadian rhythms, are orchestrated by a conserved molecular timekeeper, known as the circadian clock. The circadian clock forms a transcription-translation feedback loop that has emerged as a central biological regulator of many 24-h processes. Early studies of the intestine discovered that many digestive functions have a daily rhythm and that intestinal cell production was similarly time-dependent. As genetic methods in model organisms have become available, it has become apparent that the circadian clock regulates many basic cellular functions, including growth, proliferation, and differentiation, as well as cell signalling and stem cell self-renewal. Recent connections between circadian rhythms and immune system function, and between circadian rhythms and microbiome dynamics, have also been revealed in the intestine. These processes are highly relevant in understanding intestinal stem cell biology. Here we describe the circadian clock regulation of intestinal stem cells primarily in two model organisms: Drosophila melanogaster and mice. Like all cells in the body, intestinal stem cells are subject to circadian timing, and both cell-intrinsic and cell-extrinsic circadian processes contribute to their function.
Collapse
Affiliation(s)
- Kathyani Parasram
- Department of Biological Sciences, University of Windsor, 401 Sunset Avenue, Windsor, ON, N9B 3P4, Canada
| | - Phillip Karpowicz
- Department of Biological Sciences, University of Windsor, 401 Sunset Avenue, Windsor, ON, N9B 3P4, Canada.
| |
Collapse
|
33
|
Adamiak M, Ciechanowicz A, Skoda M, Cymer M, Tracz M, Xu B, Ratajczak MZ. Novel Evidence that Purinergic Signaling - Nlrp3 Inflammasome Axis Regulates Circadian Rhythm of Hematopoietic Stem/Progenitor Cells Circulation in Peripheral Blood. Stem Cell Rev Rep 2020; 16:335-343. [PMID: 31939051 PMCID: PMC7152586 DOI: 10.1007/s12015-020-09953-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We found that circadian changes in ATP level in peripheral blood (PB) activate the Nlrp3 inflammasome, which triggers diurnal release of hematopoietic stem/progenitor cells (HSPCs) from murine bone marrow (BM) into PB. Consistent with this finding, we observed circadian changes in expression of mRNA for Nlrp3 inflammasome-related genes, including Nlrp3, caspase 1, IL-1β, IL-18, gasdermin (GSDMD), HMGB1, and S100A9. Circadian release of HSPCs from BM into PB as well as expression of Nlrp3-associated genes was decreased in mice in which pannexin 1-mediated secretion of ATP was inhibited by the blocking peptide 10Panx and in animals exposed to the specific small-molecule inhibitor of the Nlrp3 inflammasome MCC950. In addition to HSPCs, a similar decrease in diurnal cell counts was observed for mesenchymal stromal cells (MSCs), endothelial progenitor cells (EPCs), and very small embryonic-like stem cells (VSELs). These results shed more light on the complexity of circadian regulation of HSPC release into PB, which is coordinated in a purinergic signaling-, innate immunity-dependent manner. Moreover, in addition to circadian changes in expression of the Nlrp3 inflammasome we also observed diurnal changes in expression of other inflammasomes, including Aim2, Nrp1a, and Nlrp1b.
Collapse
Affiliation(s)
- Mateusz Adamiak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Andrzej Ciechanowicz
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Marta Skoda
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Monika Cymer
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Michal Tracz
- Institute of Veterinary Medicine, Department of Food Hygiene and Public Health Protection, Warsaw University of Life Sciences, Warsaw, Poland
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology of Xiamen University, Xiamen, People’s Republic of China
| | - Mariusz Z. Ratajczak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| |
Collapse
|
34
|
Okawa H, Egusa H, Nishimura I. Implications of the circadian clock in implant dentistry. Dent Mater J 2020; 39:173-180. [PMID: 32115492 DOI: 10.4012/dmj.2019-291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Circadian rhythms are approximately 24-h cell-autonomous cycles driven by transcription and translation feedback loops of a set of core circadian clock genes, such as circadian locomoter output cycles kaput (Clock), brain and muscle arnt-like protein-1 (Bmal1), period (Per), and cryptochrome (Cry). The genetic clockwork of these genes produces circadian rhythms in cells throughout the body, including the craniofacial region. During development, dento-alveolar bone tissue formation could be regulated by site-specific circadian patterns. Studies using knockout mice and mesenchymal stem cells (MSCs) to evaluate clock genes revealed regulatory effects of clock function on bone remodeling, suggesting involvement of the circadian clockwork in osseointegration of titanium implants. Indeed, rough surface titanium modulates specific clock genes, Neuronal PAS domain protein-2 (Npas2) and Per, in MSCs to facilitate osseointegration. Further understanding of the bone clock machinery associated with biomaterial surface properties might improve preoperative diagnosis for dental implant treatments.
Collapse
Affiliation(s)
- Hiroko Okawa
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry.,Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry
| | - Ichiro Nishimura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry
| |
Collapse
|
35
|
Tsuchiya Y, Umemura Y, Yagita K. Circadian clock and cancer: From a viewpoint of cellular differentiation. Int J Urol 2020; 27:518-524. [PMID: 32223039 DOI: 10.1111/iju.14231] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/27/2020] [Indexed: 12/18/2022]
Abstract
The circadian clock controls and adapts diverse physiological and behavioral processes according to Earth's 24-h cycle of environmental changes. The master pacemaker of the mammalian circadian clock resides in the hypothalamic suprachiasmatic nucleus, but almost all cells throughout the body show circadian oscillations in gene expression patterns and associated functions. Recent studies have shown that the circadian clock gradually develops during embryogenesis. Embryonic stem cells and induced pluripotent stem cells do not show circadian oscillations of gene expression, but gradually develop circadian clock oscillation during differentiation; thus, the developmental program of circadian clock emergence appears closely associated with cellular differentiation. Like embryonic stem cells, certain cancer cell types also lack the circadian clock. Given this similarity between embryonic stem cells and cancer cells, interest is growing in the contributions of circadian clock dysfunction to dedifferentiation and cancer development. In this review, we summarize recent advances in our understanding of circadian clock emergence during ontogenesis, and discuss possible associations with cellular differentiation and carcinogenesis. Considering the multiple physiological functions of circadian rhythms, circadian abnormalities might contribute to a host of diseases, including cancer. Insights on circadian function could lead to the identification of biomarkers for cancer diagnosis and prognosis, as well as novel targets for treatment.
Collapse
Affiliation(s)
- Yoshiki Tsuchiya
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiro Umemura
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
36
|
Arafa K, Emara M. Insights About Circadian Clock and Molecular Pathogenesis in Gliomas. Front Oncol 2020; 10:199. [PMID: 32195174 PMCID: PMC7061216 DOI: 10.3389/fonc.2020.00199] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/05/2020] [Indexed: 12/15/2022] Open
Abstract
The circadian clock is an endogenous time-keeping system that has been discovered across kingdoms of life. It controls and coordinates metabolism, physiology, and behavior to adapt to variations within the day and the seasonal environmental cycles driven by earth rotation. In mammals, although circadian rhythm is controlled by a set of core clock genes that are present in both in suprachiasmatic nucleus (SCN) of the hypothalamus and peripheral tissues, the generation and control of the circadian rhythm at the cellular, tissue, and organism levels occurs in a hierarchal fashion. The SCN is central pacemaker comprising the principal circadian clock that synchronizes peripheral circadian clocks to their appropriate phase. Different epidemiological studies have shown that disruption of normal circadian rhythm is implicated in increasing the risk of developing cancers. In addition, deregulated expression of clock genes has been demonstrated in various types of cancer. These findings indicate a close association between circadian clock and cancer development and progression. Here, we review different evidences of this association in relation to molecular pathogenesis in gliomas.
Collapse
Affiliation(s)
| | - Marwan Emara
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Cairo, Egypt
| |
Collapse
|
37
|
Ullah M, Ng NN, Concepcion W, Thakor AS. Emerging role of stem cell-derived extracellular microRNAs in age-associated human diseases and in different therapies of longevity. Ageing Res Rev 2020; 57:100979. [PMID: 31704472 DOI: 10.1016/j.arr.2019.100979] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
Organismal aging involves the progressive decline in organ function and increased susceptibility to age-associated diseases. This has been associated with the aging of stem cell populations within the body that decreases the capacity of stem cells to self-renew, differentiate, and regenerate damaged tissues and organs. This review aims to explore how aging is associated with the dysregulation of stem cell-derived extracellular vesicles (SCEVs) and their corresponding miRNA cargo (SCEV-miRNAs), which are short non-coding RNAs involved in post-transcriptional regulation of target genes. Recent evidence has suggested that in aging stem cells, SCEV-miRNAs may play a vital role regulating various processes that contribute to aging: cellular senescence, stem cell exhaustion, telomere length, and circadian rhythm. Hence, further clarifying the age-dependent molecular mechanisms through which SCEV-miRNAs exert their downstream effects may inform a greater understanding of the biology of aging, elucidate their role in stem cell function, and identify important targets for future regenerative therapies. Additionally, current studies evaluating therapeutic role of SCEVs and SCEV-miRNAs in treating several age-associated diseases are also discussed.
Collapse
Affiliation(s)
- Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA.
| | - Nathan Norton Ng
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA
| | - Waldo Concepcion
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California, 94304, USA
| |
Collapse
|
38
|
Intestinal Stem Cells Exhibit Conditional Circadian Clock Function. Stem Cell Reports 2019; 11:1287-1301. [PMID: 30428387 PMCID: PMC6235668 DOI: 10.1016/j.stemcr.2018.10.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
The circadian clock is a molecular pacemaker that produces 24-hr physiological cycles known as circadian rhythms. How the clock regulates stem cells is an emerging area of research with many outstanding questions. We tested clock function in vivo at the single cell resolution in the Drosophila intestine, a tissue that is exquisitely sensitive to environmental cues and has circadian rhythms in regeneration. Our results indicate that circadian clocks function in intestinal stem cells and enterocytes but are downregulated during enteroendocrine cell differentiation. Drosophila intestinal cells are principally synchronized by the photoperiod, but intestinal stem cell clocks are highly responsive to signaling pathways that comprise their niche, and we find that the Wnt and Hippo signaling pathways positively regulate stem cell circadian clock function. These data reveal that intestinal stem cell circadian rhythms are regulated by cellular signaling and provide insight as to how clocks may be altered during physiological changes such as regeneration and aging. Intestinal epithelial cells have circadian clock function but enteroendocrine cells do not Restricted feeding can entrain circadian clocks in the absence of photoperiod Circadian clock communication exists between intestinal stem cells and enterocytes Notch, Wnt, and Hippo signaling regulate stem cell clock function
Collapse
|
39
|
Bild A, Teo JL, Kahn M. Enhanced Kat3A/Catenin transcription: a common mechanism of therapeutic resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:917-932. [PMID: 32426696 PMCID: PMC7234864 DOI: 10.20517/cdr.2019.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/04/2019] [Accepted: 06/18/2019] [Indexed: 11/12/2022]
Abstract
Cancers are heterogeneous at the cellular level. Cancer stem cells/tumor initiating cells (CSC/TIC) both initiate tumorigenesis and are responsible for therapeutic resistance and disease relapse. Elimination of CSC/TIC should therefore be able to reverse therapy resistance. In principle, this could be accomplished by either targeting cancer stem cell surface markers or "stemness" pathways. Although the successful therapeutic elimination of "cancer stemness" is a critical goal, it is complex in that it should be achieved without depletion of or increases in somatic mutations in normal tissue stem cell populations. In this perspective, we will discuss the prospects for this goal via pharmacologically targeting differential Kat3 coactivator/Catenin usage, a fundamental transcriptional control mechanism in stem cell biology.
Collapse
Affiliation(s)
- Andrea Bild
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Jia-Ling Teo
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Michael Kahn
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
40
|
Yin and Yang: Why did evolution implement and preserve the circadian rhythmicity? Med Hypotheses 2019; 131:109306. [PMID: 31443763 DOI: 10.1016/j.mehy.2019.109306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/15/2019] [Accepted: 07/06/2019] [Indexed: 02/07/2023]
Abstract
Yin and Yang concept emphasizes the reciprocal and interrelated nature; neither is sufficient, both are needed to sustain the overall balance of the living system. Changing the balance, by implementing deficiency or excess of one of them, upsets the equilibrium (homeostasis) of the whole system. PURPOSE In this opinion article intermittent exposure is presented as the stimulus for development and evolutionary conservation of circadian rhythm, an endogenous, entrainable oscillation of approximately 24 h, to counteract/balance the cells' natural tendency to attenuate their response during long-term exposure to different endogenous substances. RESULTS The concept of Yin and Yang duality is an allegory on which the avoidance of attenuation of the cells' responses hypothesis is presented as an explanation for the circadian rhythmicity, which is integrated in all human cells, with the exception of stem and cancer cells. CONCLUSIONS We hypothesize, that circadian rhythmicity has evolved, during evolution, into a mechanism that prevents disruption of the organism's negative-feedback-loop homeostasis.
Collapse
|
41
|
Avishai E, Golubnitschaja O. Flammer Syndrome in the Context of Healing Impairments – Facts and Hypotheses for Multi-professional Consideration. FLAMMER SYNDROME 2019. [DOI: 10.1007/978-3-030-13550-8_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Sun Y, Wang P, Li H, Dai J. BMAL1 and CLOCK proteins in regulating UVB-induced apoptosis and DNA damage responses in human keratinocytes. J Cell Physiol 2018; 233:9563-9574. [PMID: 29943823 PMCID: PMC6185778 DOI: 10.1002/jcp.26859] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022]
Abstract
A diverse array of biological processes are under circadian controls. In mouse skin, ultraviolet ray (UVR)-induced apoptosis and DNA damage responses are time-of-day dependent, which are controlled by core clock proteins. This study investigates the roles of clock proteins in regulating UVB responses in human keratinocytes (HKCs). We found that the messenger RNA expression of brain and muscle ARNT-like 1 (BMAL1) and circadian locomotor output cycles kaput (CLOCK) genes is altered by low doses (5 mJ/cm2 ) of UVB in the immortalized HaCat HKCs cell line. Although depletion of BMAL1 or CLOCK has no effect on the activation of Rad3-related protein kinases-checkpoint kinase 1-p53 mediated DNA damage checkpoints, it leads to suppression of UVB-stimulated apoptotic responses, and downregulation of UVB-elevated expression of DNA damage marker γ-H2AX and cell cycle inhibitor p21. Diminished apoptotic responses are also observed in primary HKCs depleted of BMAL1 or CLOCK after UVB irradiation. While CLOCK depletion shows a suppressive effect on UVB-induced p53 protein accumulation, depletion of either clock gene triggers early keratinocyte differentiation of HKCs at their steady state. These results suggest that UVB-induced apoptosis and DNA damage responses are controlled by clock proteins, but via different mechanisms in the immortalized human adult low calcium temperature and primary HKCs. Given the implication of UVB in photoaging and photocarcinogenesis, mechanistic elucidation of circadian controls on UVB effects in human skin will be critical and beneficial for prevention and treatment of skin cancers and other skin-related diseases.
Collapse
Affiliation(s)
- Yang Sun
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
| | - Peiling Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, WI 53705 USA
| | - Hongyu Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
| | - Jun Dai
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, WI 53705 USA
| |
Collapse
|
43
|
Rong J, Zhu M, Munro J, Cornish J, McCarthy GM, Dalbeth N, Poulsen RC. Altered expression of the core circadian clock component PERIOD2 contributes to osteoarthritis-like changes in chondrocyte activity. Chronobiol Int 2018; 36:319-331. [PMID: 30403881 DOI: 10.1080/07420528.2018.1540493] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In osteoarthritis, chondrocytes undergo a phenotype shift characterised by reduced expression of SOX9 (sry-box 9) and increased production of cartilage-degrading enzymes, e.g. MMP13 (matrix metalloproteinase 13) and ADAMTS5 (a disintegrin and metalloproteinase with thrombospondin motifs 5). The chondrocyte clock is also altered. Specifically, the peak level of PER2 is elevated, but peak level of BMAL1 reduced in osteoarthritic chondrocytes. The purpose of this study was to determine whether increased PER2 expression causes disease-associated changes in chondrocyte activity and to identify whether known risk factors for osteoarthritis induce changes in PER2 and BMAL1 expression. Primary human chondrocytes isolated from macroscopically normal cartilage were serum-starved overnight then re-fed with serum-replete media with/without interleukin 1β (IL-1β) (10 ng/mL), hydrogen peroxide (100 µM) or basic calcium phosphate (BCP) crystals (50 µg/mL). Peak level of BMAL1 was lower, whereas PER2 levels remained elevated for longer, in chondrocytes treated with IL-1β, hydrogen peroxide or BCP crystals compared to untreated cells. Levels of SOX9 were lower, whereas levels of ADAMTS5 and MMP13 were higher, in chondrocytes exposed to any of the three treatments compared to untreated cells. Knockdown of PER2 using siRNA partially abrogated the effects of each treatment on chondrocyte phenotype marker expression. Similarly, in chondrocytes isolated from osteoarthritic cartilage PER2 knockdown was associated with increased SOX9, reduced ADAMTS5 and reduced RNA and protein levels of MMP13 indicating partial mitigation of the osteoarthritic phenotype. Conversely, further ablation of BMAL1 expression in osteoarthritic chondrocytes resulted in a further reduction in SOX9 and increase in MMP13 expression. Overexpression of PER2 in the H5 chondrocyte cell line led to increased ADAMTS5 and MMP13 and decreased SOX9 expression. Localised inflammation, oxidative stress and BCP crystal deposition in osteoarthritic joints may contribute to disease pathology by inducing changes in the chondrocyte circadian clock.
Collapse
Affiliation(s)
- Jing Rong
- a Department of Medicine , School of Medicine, University of Auckland , Auckland , New Zealand
| | - Mark Zhu
- a Department of Medicine , School of Medicine, University of Auckland , Auckland , New Zealand.,b Department of Surgery, School of Medicine , University of Auckland , Auckland , New Zealand
| | - Jacob Munro
- b Department of Surgery, School of Medicine , University of Auckland , Auckland , New Zealand
| | - Jillian Cornish
- a Department of Medicine , School of Medicine, University of Auckland , Auckland , New Zealand
| | | | - Nicola Dalbeth
- a Department of Medicine , School of Medicine, University of Auckland , Auckland , New Zealand
| | - Raewyn C Poulsen
- a Department of Medicine , School of Medicine, University of Auckland , Auckland , New Zealand
| |
Collapse
|
44
|
Kalev-Zylinska ML, Hearn JI, Rong J, Zhu M, Munro J, Cornish J, Dalbeth N, Poulsen RC. Altered N-methyl D-aspartate receptor subunit expression causes changes to the circadian clock and cell phenotype in osteoarthritic chondrocytes. Osteoarthritis Cartilage 2018; 26:1518-1530. [PMID: 30031924 DOI: 10.1016/j.joca.2018.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/08/2018] [Accepted: 06/30/2018] [Indexed: 02/02/2023]
Abstract
UNLABELLED The chondrocyte circadian clock is altered in osteoarthritis. This change is implicated in the disease-associated changes in chondrocyte phenotype and cartilage loss. Why the clock is changed is unknown. N-methyl-D-aspartate receptors (NMDAR) are critical for regulating the hypothalamic clock. Chondrocytes also express NMDAR and the type of NMDAR subunits expressed changes in osteoarthritis. OBJECTIVE To determine if NMDAR regulate the chondrocyte clock and phenotype. DESIGN Chondrocytes isolated from macroscopically-normal (MN) and osteoarthritic human cartilage were treated with NMDAR antagonists or transfected with GRIN2A or GRIN2B-targetting siRNA. H5 chondrocytes were transfected with GluN2B-expression plasmids. Clock genes and chondrocyte phenotypic markers were measured by RT-qPCR. RESULTS PER2 amplitude was higher and BMAL1 amplitude lower in osteoarthritic compared to MN chondrocytes. In osteoarthritic chondrocytes, NMDAR inhibition restored PER2 and BMAL1 expression to levels similar to MN chondrocytes, and resulted in reduced MMP13 and COL10A1. Paradoxically, NMDAR inhibition in MN chondrocytes resulted in increased PER2, decreased BMAL1 and increased MMP13 and COL10A1. Osteoarthritic, but not MN chondrocytes expressed GluN2B NMDAR subunits. GluN2B knockdown in osteoarthritic chondrocytes restored expression of circadian clock components and phenotypic markers to levels similar to MN chondrocytes. Ectopic expression of GluN2B resulted in reduced BMAL1, increased PER2 and altered SOX9, RUNX2 and MMP13 expression. Knockdown of PER2 mitigated the effects of GluN2B on SOX9 and MMP13. CONCLUSIONS NMDAR regulate the chondrocyte clock and phenotype suggesting NMDAR may also regulate clocks in other peripheral tissues. GluN2B expression in osteoarthritis may contribute to pathology by altering the chondrocyte clock.
Collapse
Affiliation(s)
- M L Kalev-Zylinska
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Auckland, New Zealand.
| | - J I Hearn
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Auckland, New Zealand.
| | - J Rong
- Department of Medicine, School of Medicine, Auckland, New Zealand.
| | - M Zhu
- Department of Medicine, School of Medicine, Auckland, New Zealand; Department of Surgery, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - J Munro
- Department of Surgery, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - J Cornish
- Department of Medicine, School of Medicine, Auckland, New Zealand.
| | - N Dalbeth
- Department of Medicine, School of Medicine, Auckland, New Zealand.
| | - R C Poulsen
- Department of Medicine, School of Medicine, Auckland, New Zealand.
| |
Collapse
|
45
|
Rogers EH, Hunt JA, Pekovic-Vaughan V. Adult stem cell maintenance and tissue regeneration around the clock: do impaired stem cell clocks drive age-associated tissue degeneration? Biogerontology 2018; 19:497-517. [PMID: 30374678 PMCID: PMC6223734 DOI: 10.1007/s10522-018-9772-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023]
Abstract
Human adult stem cell research is a highly prolific area in modern tissue engineering as these cells have significant potential to provide future cellular therapies for the world's increasingly aged population. Cellular therapies require a smart biomaterial to deliver and localise the cell population; protecting and guiding the stem cells toward predetermined lineage-specific pathways. The cells, in turn, can provide protection to biomaterials and increase its longevity. The right combination of stem cells and biomaterials can significantly increase the therapeutic efficacy. Adult stem cells are utilised to target many changes that negatively impact tissue functions with age. Understanding the underlying mechanisms that lead to changes brought about by the ageing process is imperative as ageing leads to many detrimental effects on stem cell activation, maintenance and differentiation. The circadian clock is an evolutionarily conserved timing mechanism that coordinates physiology, metabolism and behavior with the 24 h solar day to provide temporal tissue homeostasis with the external environment. Circadian rhythms deteriorate with age at both the behavioural and molecular levels, leading to age-associated changes in downstream rhythmic tissue physiology in humans and rodent models. In this review, we highlight recent advances in our knowledge of the role of circadian clocks in adult stem cell maintenance, driven by both cell-autonomous and tissue-specific factors, and the mechanisms by which they co-opt various cellular signaling pathways to impose temporal control on stem cell function. Future research investigating pharmacological and lifestyle interventions by which circadian rhythms within adult stem niches can be manipulated will provide avenues for temporally guided cellular therapies and smart biomaterials to ameliorate age-related tissue deterioration and reduce the burden of chronic disease.
Collapse
Affiliation(s)
- Eve H Rogers
- Institute of Ageing and Chronic Disease, University of Liverpool, The William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - John A Hunt
- School of Science and Technology, Nottingham Trent University, Clifton Campus, College Drive, Nottingham, NG11 8NS, UK
| | - Vanja Pekovic-Vaughan
- Institute of Ageing and Chronic Disease, University of Liverpool, The William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| |
Collapse
|
46
|
Elkhenany H, AlOkda A, El-Badawy A, El-Badri N. Tissue regeneration: Impact of sleep on stem cell regenerative capacity. Life Sci 2018; 214:51-61. [PMID: 30393021 DOI: 10.1016/j.lfs.2018.10.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 12/31/2022]
Abstract
The circadian rhythm orchestrates many cellular functions, such as cell division, cell migration, metabolism and numerous intracellular biological processes. The physiological changes during sleep are believed to promote a suitable microenvironment for stem cells to proliferate, migrate and differentiate. These effects are mediated either directly by circadian clock genes or indirectly via hormones and cytokines. Hormones, such as melatonin and cortisol, are secreted in response to neural optic signals and act in harmony to regulate many biological functions during sleep. Herein, we correlate the effects of the main circadian genes on the expression of certain stem cell genes responsible for the regeneration of different tissues, including bone, cartilage, skin, and intestine. We also review the effects of different hormones and cytokines on stem cell activation or suppression and their relationship to the day/night cycle. The correlation of circadian rhythm with tissue regeneration could have implications in understanding the biology of sleep and tissue regeneration and in enhancing the efficacy and timing of surgical procedures.
Collapse
Affiliation(s)
- Hoda Elkhenany
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt; Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, 22785, Egypt
| | - Abdelrahman AlOkda
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Ahmed El-Badawy
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Nagwa El-Badri
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt.
| |
Collapse
|
47
|
Budkowska M, Ostrycharz E, Wojtowicz A, Marcinowska Z, Woźniak J, Ratajczak MZ, Dołęgowska B. A Circadian Rhythm in both Complement Cascade (ComC) Activation and Sphingosine-1-Phosphate (S1P) Levels in Human Peripheral Blood Supports a Role for the ComC-S1P Axis in Circadian Changes in the Number of Stem Cells Circulating in Peripheral Blood. Stem Cell Rev Rep 2018; 14:677-685. [PMID: 29911288 PMCID: PMC6132735 DOI: 10.1007/s12015-018-9836-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The number of hematopoietic stem/progenitor cells (HSPCs) circulating in peripheral blood (PB) is regulated by a circadian rhythm, and more HSPCs circulate in PB in the morning hours than at night. Different mechanisms have been proposed that might regulate this process, including changes in tonus of β-adrenergic innervation of bone marrow (BM) tissue. Our group reported that in mice circadian changes in the number of HSPCs circulating in PB correlates with diurnal activation of the complement cascade (ComC) and that the mice deficient in C5 component of ComC (C5-KO mice) do not show circadian changes in the number of circulating HSPCs in PB. We also reported the existence of a gradient between PB and BM of a bioactive phosphosphingolipid, sphingosine-1-phosphate (S1P), which is a major PB chemottractant for BM-residing HSPCs. Based on these observations, we investigated activation of the ComC and the level of S1P in the PB of 66 healthy volunteers. We found that both ComC activation and the S1P level undergo changes in a circadian cycle. While the ComC becomes highly activated during deep sleep at 2 am, S1P becomes activated later, and its highest level is observed at 8 am, which precedes circadian egress of HSPCs from BM into PB. In sum, circadian activation of the ComC-S1P axis releases HSPCs from BM into PB.
Collapse
Affiliation(s)
- Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland.
| | - Ewa Ostrycharz
- Department of Medical Analytics, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Adrianna Wojtowicz
- Department of Medical Analytics, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Zuzanna Marcinowska
- Centre for Human Structural and Functional Research, Faculty of Physical Education and Health Promotion, University of Szczecin, ul. Narutowicza 17C, 70-240, Szczecin, Poland
| | - Jarosław Woźniak
- Institute of Mathematics, Department of Mathematics and Physics, University of Szczecin, Ul. Wielkopolska 15, 70-451, Szczecin, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, ul. Banacha 1B, 02-097, Warsaw, Poland
| | - Barbara Dołęgowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| |
Collapse
|
48
|
Abstract
Biology is dynamic. Timescales range from frenetic sub-second ion fluxes and enzymatic reactions to the glacial millions of years of evolutionary change. Falling somewhere in the middle of this range are the processes we usually study in development: cell division and differentiation, gene expression, cell-cell signalling, and morphogenesis. But what sets the tempo and manages the order of developmental events? Are the order and tempo different between species? How is the sequence of multiple events coordinated? Here, we discuss the importance of time for developing embryos, highlighting the necessity for global as well as cell-autonomous control. New reagents and tools in imaging and genomic engineering, combined with in vitro culture, are beginning to offer fresh perspectives and molecular insight into the origin and mechanisms of developmental time.
Collapse
Affiliation(s)
- Miki Ebisuya
- RIKEN Center for Biosystems Dynamics Research (RIKEN BDR), 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - James Briscoe
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
49
|
Crnko S, Cour M, Van Laake LW, Lecour S. Vasculature on the clock: Circadian rhythm and vascular dysfunction. Vascul Pharmacol 2018; 108:1-7. [PMID: 29778521 DOI: 10.1016/j.vph.2018.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/23/2018] [Accepted: 05/10/2018] [Indexed: 01/08/2023]
Abstract
The master mammalian circadian clock (i.e. central clock), located in the suprachiasmatic nucleus of the hypothalamus, orchestrates the synchronization of the daily behavioural and physiological rhythms to better adapt the organism to the external environment in an anticipatory manner. This central clock is entrained by a variety of signals, the best established being light and food. However, circadian cycles are not simply the consequences of these two cues but are generated by endogenous circadian clocks. Indeed, clock machinery is found in mainly all tissues and cell types, including cells of the vascular system such as endothelial cells, fibroblasts, smooth muscle cells and stem cells. This machinery physiologically contributes to modulate the daily vascular function, and its disturbance therefore plays a major role in the pathophysiology of vascular dysfunction. Therapies targeting the circadian rhythm may therefore be of benefit against vascular disease.
Collapse
Affiliation(s)
- Sandra Crnko
- Division Heart and Lungs and Regenerative Medicine Center, University Medical Center Utrecht, The Netherlands
| | - Martin Cour
- Hatter Institute for Cardiovascular research in Africa and Lionel Opie Preclinical Imaging Core Facility, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Linda W Van Laake
- Division Heart and Lungs and Regenerative Medicine Center, University Medical Center Utrecht, The Netherlands
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular research in Africa and Lionel Opie Preclinical Imaging Core Facility, Faculty of Health Sciences, University of Cape Town, South Africa.
| |
Collapse
|
50
|
Rakshit K, Qian J, Gaonkar KS, Dhawan S, Colwell CS, Matveyenko AV. Postnatal Ontogenesis of the Islet Circadian Clock Plays a Contributory Role in β-Cell Maturation Process. Diabetes 2018; 67:911-922. [PMID: 29500314 PMCID: PMC5910002 DOI: 10.2337/db17-0850] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 02/23/2018] [Indexed: 12/13/2022]
Abstract
Development of cell replacement therapies in diabetes requires understanding of the molecular underpinnings of β-cell maturation. The circadian clock regulates diverse cellular functions important for regulation of β-cell function and turnover. However, postnatal ontogenesis of the islet circadian clock and its potential role in β-cell maturation remain unknown. To address this, we studied wild-type Sprague-Dawley as well as Period1 luciferase transgenic (Per1:LUC) rats to determine circadian clock function, clock protein expression, and diurnal insulin secretion during islet development and maturation process. We additionally studied β-cell-specific Bmal1-deficient mice to elucidate a potential role of this key circadian transcription factor in β-cell functional and transcriptional maturation. We report that emergence of the islet circadian clock 1) occurs during the early postnatal period, 2) depends on the establishment of global behavioral circadian rhythms, and 3) leads to the induction of diurnal insulin secretion and gene expression. Islet cell maturation was also characterized by induction in the expression of circadian transcription factor BMAL1, deletion of which altered postnatal development of glucose-stimulated insulin secretion and the associated transcriptional network. Postnatal development of the islet circadian clock contributes to early-life β-cell maturation and should be considered for optimal design of future β-cell replacement strategies in diabetes.
Collapse
Affiliation(s)
- Kuntol Rakshit
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN
| | - Jingyi Qian
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| | - Krutika Satish Gaonkar
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN
| | - Sangeeta Dhawan
- Department of Translational Research & Cellular Therapeutics, City of Hope, Duarte, CA
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|