1
|
Wang W, Yuan G, Li G, Zhao T, Chen Y, Xu Y. Cigarette smoking extract induces mitochondrial dysfunction and apoptosis in HUVECs via the Sirt1-SHH axis. Hum Exp Toxicol 2025; 44:9603271251332251. [PMID: 40170556 DOI: 10.1177/09603271251332251] [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] [Indexed: 04/03/2025]
Abstract
IntroductionCigarette smoking extract (CSE) can cause endothelial cell (EC) dysfunction, and then promote the occurrence and development of atherosclerosis. However, the molecular mechanisms underlying CSE-induced EC dysfunction are unknown. Sirt1, as a deacetylase, is involved in various biological processes of ECs. Therefore, this study investigated whether CSE induces apoptosis and mitochondrial dysfunction in human umbilical vein endothelial cells (HUVECs) via Sirt1-dependent mechanisms.MethodsHUVEC activity was assessed using MTT and crystal violet staining following treatment with different concentrations of CSE. Lentiviral transfection technology was used to generate HUVECs overexpressing Sirt1. Apoptosis was detected by Tunnel staining. MitoTracker™ Deep Red FM and JC-1 were used to assess mitochondrial structure and membrane potential. ELISA was used to detect the expression of superoxide dismutase (SOD) and malondialdehyde (MDA). qPCR was used to determine mRNA expression. Atherosclerosis was evaluated by oil red O staining in ApoE-KO mice after cigarette smoke exposure.ResultsCSE decreased Sirt1 and sonic hedgehog (SHH) expression, leading to mitochondrial dysfunction and apoptosis in HUVECs. Overexpressing Sirt1 or activating the SHH signaling pathway attenuated CSE-induced apoptosis and mitochondrial dysfunction. However, inhibiting the SHH signaling axis attenuated the protective effect of Sirt1 overexpression on CSE-induced apoptosis and mitochondrial dysfunction. In vivo studies also showed that cigarette smoke exacerbated atherosclerosis in ApoE-KO mice, downregulating Sirt1, SHH, and Gli1 expression in the aorta. Additionally, cigarette smoke increased Bax expression and decreased Bcl-2 expression in ApoE-KO mice aortas.DiscussionsSmoking can affect all stages of the atherosclerosis process, and the specific mechanism remains unclear. This study confirms that CSE can induce mitochondrial dysfunction and apoptosis of HUVECs by reducing Sirt1 expression and inhibiting SHH signaling activation. These findings provide new insights into the prevention and treatment of smoking-induced atherosclerosis.
Collapse
Affiliation(s)
- Weiming Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, China
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Yuan
- Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, China
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Tingting Zhao
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Yue Chen
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, China
| |
Collapse
|
2
|
Zhang Y, Savvidou M, Liaudanskaya V, Singh P, Fu Y, Nasreen A, Coe M, Kelly M, Snapper D, Wagner C, Gill J, Symes A, Patra A, Kaplan DL, Beheshti A, Georgakoudi I. Synergistic label-free fluorescence imaging and miRNA studies reveal dynamic human neuron-glial metabolic interactions following injury. SCIENCE ADVANCES 2024; 10:eadp1980. [PMID: 39661671 PMCID: PMC11633737 DOI: 10.1126/sciadv.adp1980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Neuron-glial cell interactions following traumatic brain injury (TBI) determine the propagation of damage and long-term neurodegeneration. Spatiotemporally heterogeneous cytosolic and mitochondrial metabolic pathways are involved, leading to challenges in developing effective diagnostics and treatments. An engineered three-dimensional brain tissue model comprising human neurons, astrocytes, and microglia is used in combination with label-free, two-photon imaging and microRNA studies to characterize metabolic interactions between glial and neuronal cells over 72 hours following impact injury. We interpret multiparametric, quantitative, optical metabolic assessments in the context of microRNA gene set analysis and identify distinct metabolic changes in neurons and glial cells. Glycolysis, nicotinamide adenine dinucleotide phosphate (reduced form) and glutathione synthesis, fatty acid synthesis, and oxidation are mobilized within glial cells to mitigate the impacts of initial enhancements in oxidative phosphorylation and fatty acid oxidation within neurons, which lack robust antioxidant defenses. This platform enables enhanced understanding of mechanisms that may be targeted to improve TBI diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Maria Savvidou
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Volha Liaudanskaya
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Pramesh Singh
- Data Intensive Studies Center, Tufts University, Medford, MA 02155, USA
| | - Yuhang Fu
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Amreen Nasreen
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Marly Coe
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Marilyn Kelly
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Dustin Snapper
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Chelsea Wagner
- School of Nursing, Johns Hopkins University, 525 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Jessica Gill
- School of Nursing, Johns Hopkins University, 525 N. Wolfe Street, Baltimore, MD 21205, USA
- Department of Neurology, School of Medicine, Johns Hopkins University, 525 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Aviva Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Abani Patra
- Data Intensive Studies Center, Tufts University, Medford, MA 02155, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Afshin Beheshti
- McGowan Institute for Regenerative Medicine - Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Program in Cell, Molecular, and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Dartmouth Cancer Center, Dartmouth Hitchcock Medical Center, Lebanon NH 03766, USA
| |
Collapse
|
3
|
Shweta, Sharma K, Shakarad M, Agrawal N, Maurya SK. Drosophila glial system: an approach towards understanding molecular complexity of neurodegenerative diseases. Mol Biol Rep 2024; 51:1146. [PMID: 39532789 DOI: 10.1007/s11033-024-10075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Glia is pivotal in regulating neuronal stem cell proliferation, functioning, and nervous system homeostasis, significantly influencing neuronal health and disorders. Dysfunction in glial activity is a key factor in the development and progression of brain pathology. However, a deeper understanding of the intricate nature of glial cells and their diverse role in neurological disorders is still required. To this end, we conducted data mining to retrieve literature from PubMed and Google Scholar using the keywords: glia, Drosophila, neurodegeneration, and mammals. The retrieved literature was manually screened and used to comprehensively understand and present the different glial types in Drosophila, i.e., perineurial, subperineurial, cortex, astrocyte-like and ensheathing glia, their relevance with mammalian counterparts, mainly microglia and astrocytes, and their potential to reveal complex neuron-glial molecular networks in managing neurodegenerative processes.
Collapse
Affiliation(s)
- Shweta
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Khushboo Sharma
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallikarjun Shakarad
- Evolutionary Biology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Namita Agrawal
- Fly Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
4
|
Levy I, Arvidson R. Cephalic ganglia transcriptomics of the American cockroach Periplaneta americana (Blattodea: Blattidae). JOURNAL OF INSECT SCIENCE (ONLINE) 2024; 24:12. [PMID: 39688382 DOI: 10.1093/jisesa/ieae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024]
Abstract
The American cockroach Periplaneta americana (L.) (Blattodea, Blattidae) has been a model organism for biochemical and physiological study for almost a century, however, its use does not benefit from the genetic tools found in key model species such as Drosophila melanogaster. To facilitate the use of the cockroach as a model system in neuroscience and to serve as a foundation for functional and translational experimentation, a transcriptome of the cephalic ganglia was assembled and annotated, and differential expression profiles between these ganglia were assessed. The transcriptome assembly yielded >400 k transcripts, with >40 k putative coding sequences. Gene ontology and protein domain searches indicate the cerebral and gnathal ganglia (GNG) have distinct genetic expression profiles. The developmental Toll signaling pathway appears to be active in the adult central nervous system (CNS), which may suggest a separate role for this pathway besides innate immune activation or embryonic development. The catabolic glycolytic and citric acid cycle enzymes are well represented in both ganglia, but key enzymes are more highly expressed in the GNG. Both ganglia express gluconeogenic and trehaloneogenic enzymes, suggesting a larger role of the CNS in regulating hemolymph sugar homeostasis than previously appreciated. The annotation and quantification of the cephalic ganglia transcriptome reveal both canonical and novel pathways in signaling and metabolism in an adult insect and lay a foundation for future functional and genetic analysis.
Collapse
Affiliation(s)
- Ilana Levy
- Undergraduate Program in Biochemistry, Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Ryan Arvidson
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
5
|
Palmer EM, Snoddy CA, York PM, Davis SM, Hunter MF, Krishnan N. Enhanced Age-Dependent Motor Impairment in Males of Drosophila melanogaster Modeling Spinocerebellar Ataxia Type 1 Is Linked to Dysregulation of a Matrix Metalloproteinase. BIOLOGY 2024; 13:854. [PMID: 39596808 PMCID: PMC11591802 DOI: 10.3390/biology13110854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Over the past two decades, Drosophila melanogaster has proven to be successful in modeling the polyglutamine (polyQ) (caused by CAG repeats) family of neurodegenerative disorders, including the faithful recapitulation of pathological features such as polyQ length-dependent formation of protein aggregates and progressive neuronal degeneration. In this study, pan-neuronal expression of human Ataxin-1 with long polyQ repeat of 82 amino acids was driven using an elav-GAL4 driver line. This would essentially model the polyQ disease spinocerebellar ataxia type 1 (SCA1). Longevity and behavioral analysis of male flies expressing human Ataxin-1 revealed compromised lifespan and accelerated locomotor activity deficits both in diurnal activity and negative geotaxis response compared to control flies. Interestingly, this decline in motor response was coupled to an enhancement of matrix metalloproteinase 1 (dMMP1) expression together with declining expression of extracellular matrix (ECM) fibroblast growth factor (FGF) signaling by hedgehog (Hh) and branchless (bnl) and a significant decrease in expression of survival motor neuron gene (dsmn) in old (30 d) flies. Taken together, our results indicate a role for dysregulation of matrix metalloproteinase in polyQ disease with consequent impact on ECM signaling factors, as well as SMN at the neuromuscular junction causing overt physiological and behavioral deficits.
Collapse
Affiliation(s)
| | | | | | | | | | - Natraj Krishnan
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, Mississippi State, MS 39762, USA
| |
Collapse
|
6
|
Urushihata T, Satoh A. Role of the central nervous system in cell non-autonomous signaling mechanisms of aging and longevity in mammals. J Physiol Sci 2024; 74:40. [PMID: 39217308 PMCID: PMC11365208 DOI: 10.1186/s12576-024-00934-3] [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: 03/01/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Multiple organs orchestrate the maintenance of proper physiological function in organisms throughout their lifetimes. Recent studies have uncovered that aging and longevity are regulated by cell non-autonomous signaling mechanisms in several organisms. In the brain, particularly in the hypothalamus, aging and longevity are regulated by such cell non-autonomous signaling mechanisms. Several hypothalamic neurons have been identified as regulators of mammalian longevity, and manipulating them promotes lifespan extension or shortens the lifespan in rodent models. The hypothalamic structure and function are evolutionally highly conserved across species. Thus, elucidation of hypothalamic function during the aging process will shed some light on the mechanisms of aging and longevity and, thereby benefiting to human health.
Collapse
Affiliation(s)
- Takuya Urushihata
- Department of Integrative Physiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
- Department of Integrative Physiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Akiko Satoh
- Department of Integrative Physiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
- Department of Integrative Physiology, National Center for Geriatrics and Gerontology, Obu, Japan.
| |
Collapse
|
7
|
Jun JH, Du K, Dutta RK, Maeso-Diaz R, Oh SH, Wang L, Gao G, Ferreira A, Hill J, Pullen SS, Diehl AM. The senescence-associated secretome of Hedgehog-deficient hepatocytes drives MASLD progression. J Clin Invest 2024; 134:e180310. [PMID: 39190624 PMCID: PMC11444248 DOI: 10.1172/jci180310] [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: 02/13/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
The burden of senescent hepatocytes correlates with the severity of metabolic dysfunction-associated steatotic liver disease (MASLD), but the mechanisms driving senescence and how it exacerbates MASLD are poorly understood. Hepatocytes experience lipotoxicity and become senescent when Smoothened (Smo) is deleted to disrupt Hedgehog signaling. We aimed to determine whether the secretomes of Smo-deficient hepatocytes perpetuate senescence to drive MASLD progression. RNA-Seq analysis of liver samples from human and murine cohorts with MASLD confirmed that hepatocyte populations in MASLD livers were depleted of Smo+ cells and enriched with senescent cells. When fed a choline-deficient, amino acid-restricted high-fat diet (CDA-HFD) to induce MASLD, Smo- mice had lower antioxidant markers and developed worse DNA damage, senescence, steatohepatitis, and fibrosis than did Smo+ mice. Sera and hepatocyte-conditioned medium from Smo- mice were depleted of thymidine phosphorylase (TP), a protein that maintains mitochondrial fitness. Treating Smo- hepatocytes with TP reduced senescence and lipotoxicity, whereas inhibiting TP in Smo+ hepatocytes had the opposite effect and exacerbated hepatocyte senescence, steatohepatitis, and fibrosis in CDA-HFD-fed mice. We conclude that inhibition of Hedgehog signaling in hepatocytes promoted MASLD by suppressing hepatocyte production of proteins that prevent lipotoxicity and senescence.
Collapse
Affiliation(s)
- Ji Hye Jun
- Division of Gastroenterology, Department of Medicine and
| | - Kuo Du
- Division of Gastroenterology, Department of Medicine and
| | | | | | - Seh Hoon Oh
- Division of Gastroenterology, Department of Medicine and
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Guannan Gao
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Ana Ferreira
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Jon Hill
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Steven S Pullen
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine and
| |
Collapse
|
8
|
Ramasubbu K, Venkatraman G, Ramanathan G, Dhanasekar S, Rajeswari VD. Molecular and cellular signalling pathways for promoting neural tissue growth - A tissue engineering approach. Life Sci 2024; 346:122640. [PMID: 38614302 DOI: 10.1016/j.lfs.2024.122640] [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: 02/08/2024] [Revised: 03/18/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Neural tissue engineering is a sub-field of tissue engineering that develops neural tissue. Damaged central and peripheral nervous tissue can be fabricated with a suitable scaffold printed with biomaterials. These scaffolds promote cell growth, development, and migration, yet they vary according to the biomaterial and scaffold printing technique, which determine the physical and biochemical properties. The physical and biochemical properties of scaffolds stimulate diverse signalling pathways, such as Wnt, NOTCH, Hedgehog, and ion channels- mediated pathways to promote neuron migration, elongation and migration. However, neurotransmitters like dopamine, acetylcholine, gamma amino butyric acid, and other signalling molecules are critical in neural tissue engineering to tissue fabrication. Thus, this review focuses on neural tissue regeneration with a tissue engineering approach highlighting the signalling pathways. Further, it explores the interaction of the scaffolds with the signalling pathways for generating neural tissue.
Collapse
Affiliation(s)
- Kanagavalli Ramasubbu
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Ganasambanthan Ramanathan
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Sivaraman Dhanasekar
- Department of Biotechnology, Pandit Deendayal Energy University, Gandhinagar 382007, Gujarat, India
| | - V Devi Rajeswari
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
9
|
Pires da Silva A, Kelleher R, Reynoldson L. Decoding lifespan secrets: the role of the gonad in Caenorhabditis elegans aging. FRONTIERS IN AGING 2024; 5:1380016. [PMID: 38605866 PMCID: PMC11008531 DOI: 10.3389/fragi.2024.1380016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024]
Abstract
The gonad has become a central organ for understanding aging in C. elegans, as removing the proliferating stem cells in the germline results in significant lifespan extension. Similarly, when starvation in late larval stages leads to the quiescence of germline stem cells the adult nematode enters reproductive diapause, associated with an extended lifespan. This review summarizes recent advancements in identifying the mechanisms behind gonad-mediated lifespan extension, including comparisons with other nematodes and the role of lipid signaling and transcriptional changes. Given that the gonad also mediates lifespan regulation in other invertebrates and vertebrates, elucidating the underlying mechanisms may help to gain new insights into the mechanisms and evolution of aging.
Collapse
|
10
|
Dutta RK, Jun J, Du K, Diehl AM. Hedgehog Signaling: Implications in Liver Pathophysiology. Semin Liver Dis 2023; 43:418-428. [PMID: 37802119 DOI: 10.1055/a-2187-3382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
The purpose of this review is to summarize current knowledge about the role of the Hedgehog signaling pathway in liver homeostasis and disease. Hedgehog is a morphogenic signaling pathway that is active in development. In most healthy tissues, pathway activity is restricted to stem and/or stromal cell compartments, where it enables stem cell self-renewal and tissue homeostasis. Aberrant over-activation of Hedgehog signaling occurs in many cancers, including hepatocellular and cholangio-carcinoma. The pathway is also activated transiently in stromal cells of injured tissues and orchestrates normal wound healing responses, including inflammation, vascular remodeling, and fibrogenesis. In liver, sustained Hedgehog signaling in stromal cells plays a major role in the pathogenesis of cirrhosis. Hedgehog signaling was thought to be silenced in healthy hepatocytes. However, recent studies show that targeted disruption of the pathway in hepatocytes dysregulates lipid, cholesterol, and bile acid metabolism, and promotes hepatic lipotoxicity, insulin resistance, and senescence. Hepatocytes that lack Hedgehog activity also produce a secretome that activates Hedgehog signaling in cholangiocytes and neighboring stromal cells to induce inflammatory and fibrogenic wound healing responses that drive progressive fibrosis. In conclusion, Hedgehog signaling must be precisely controlled in adult liver cells to maintain liver health.
Collapse
Affiliation(s)
| | - JiHye Jun
- Department of Medicine, Duke University, Durham, North Carolina
| | - Kuo Du
- Department of Medicine, Duke University, Durham, North Carolina
| | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
11
|
Shi C, Murphy CT. piRNAs regulate a Hedgehog germline-to-soma pro-aging signal. NATURE AGING 2023; 3:47-63. [PMID: 37118518 PMCID: PMC10154208 DOI: 10.1038/s43587-022-00329-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/03/2022] [Indexed: 04/30/2023]
Abstract
The reproductive system regulates somatic aging through competing anti- and pro-aging signals. Germline removal extends somatic lifespan through conserved pathways including insulin and mammalian target-of-rapamycin signaling, while germline hyperactivity shortens lifespan through unknown mechanisms. Here we show that mating-induced germline hyperactivity downregulates piRNAs, in turn desilencing their targets, including the Hedgehog-like ligand-encoding genes wrt-1 and wrt-10, ultimately causing somatic collapse and death. Germline-produced Hedgehog signals require PTR-6 and PTR-16 receptors for mating-induced shrinking and death. Our results reveal an unconventional role of the piRNA pathway in transcriptional regulation of Hedgehog signaling and a new role of Hedgehog signaling in the regulation of longevity and somatic maintenance: Hedgehog signaling is controlled by the tunable piRNA pathway to encode the previously unknown germline-to-soma pro-aging signal. Mating-induced piRNA downregulation in the germline and subsequent Hedgehog signaling to the soma enable the animal to tune somatic resource allocation to germline needs, optimizing reproductive timing and survival.
Collapse
Affiliation(s)
- Cheng Shi
- Department of Molecular Biology and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Biological Sciences, University of New Orleans, New Orleans, LA, USA.
| | - Coleen T Murphy
- Department of Molecular Biology and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
12
|
Targeting the "hallmarks of aging" to slow aging and treat age-related disease: fact or fiction? Mol Psychiatry 2023; 28:242-255. [PMID: 35840801 PMCID: PMC9812785 DOI: 10.1038/s41380-022-01680-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023]
Abstract
Aging is a major risk factor for a number of chronic diseases, including neurodegenerative and cerebrovascular disorders. Aging processes have therefore been discussed as potential targets for the development of novel and broadly effective preventatives or therapeutics for age-related diseases, including those affecting the brain. Mechanisms thought to contribute to aging have been summarized under the term the "hallmarks of aging" and include a loss of proteostasis, mitochondrial dysfunction, altered nutrient sensing, telomere attrition, genomic instability, cellular senescence, stem cell exhaustion, epigenetic alterations and altered intercellular communication. We here examine key claims about the "hallmarks of aging". Our analysis reveals important weaknesses that preclude strong and definitive conclusions concerning a possible role of these processes in shaping organismal aging rate. Significant ambiguity arises from the overreliance on lifespan as a proxy marker for aging, the use of models with unclear relevance for organismal aging, and the use of study designs that do not allow to properly estimate intervention effects on aging rate. We also discuss future research directions that should be taken to clarify if and to what extent putative aging regulators do in fact interact with aging. These include multidimensional analytical frameworks as well as designs that facilitate the proper assessment of intervention effects on aging rate.
Collapse
|
13
|
Mo L, Li R, He C, Chen Q, Xu C, Shen L, Chen K, Wu Y. Hedgehog pathway is negatively regulated during the development of Drosophila melanogaster PheRS-m (Drosophila homologs gene of human FARS2) mutants. Hum Cell 2023; 36:121-131. [PMID: 36205831 DOI: 10.1007/s13577-022-00796-0] [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: 07/20/2022] [Accepted: 09/17/2022] [Indexed: 01/20/2023]
Abstract
Hereditary spastic paraplegia (HSP) is a neurodegeneration disease, one of the reasons is caused by autosomal recessive missense mutation of the karyogene that encodes phenylalanyl-tRNA synthetase 2, mitochondrial (FARS2). However, the molecular mechanism underlying FARS2-mediated HSP progression is unknown. Mitochondrial phenylalanyl-tRNA synthetase gene (PheRS-m) is the Drosophila melanogaster homolog gene of human FARS2. This study constructed a Drosophila HSP missense mutation model and a PheRS-m knockout model. Some of the mutant fly phenotypes included developmental delay, shortened lifespan, wing-structure abnormalities and decreased mobility. RNA-sequencing results revealed a relationship between abnormal phenotypes and the hedgehog (Hh) pathway. A qRT-PCR assay was used to determine the key genes (ptc, hib, and slmb) of the Hh pathway that exhibited increased expression during different developmental stages. We demonstrated that Hh signaling transduction is negatively regulated during the developmental stages of PheRS-m mutants but positively regulated during adulthood. By inducing the agonist and inhibitor of Hh pathway in PheRS-m larvae, the developmental delay in mutants can be partly salvaged or postponed. Collectively, our findings indicate that Hh signaling negatively regulates the development of PheRS-m mutants, subsequently leading to developmental delay.
Collapse
Affiliation(s)
- Lidangzhi Mo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
- Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Rui Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
- Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chunxia He
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic & Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Qi Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
- Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Changwei Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
- Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Liangliang Shen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China.
- Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China.
| | - Kun Chen
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, School of Basic Medicine, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China.
| | - Yuanming Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China.
- Shaanxi Provincial Key Laboratory of Clinic Genetics, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
14
|
Ciulla DA, Dranchak P, Pezzullo JL, Mancusi RA, Psaras AM, Rai G, Giner JL, Inglese J, Callahan BP. A cell-based bioluminescence reporter assay of human Sonic Hedgehog protein autoprocessing to identify inhibitors and activators. J Biol Chem 2022; 298:102705. [PMID: 36400200 PMCID: PMC9772569 DOI: 10.1016/j.jbc.2022.102705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
The Sonic Hedgehog (SHh) precursor protein undergoes biosynthetic autoprocessing to cleave off and covalently attach cholesterol to the SHh signaling ligand, a vital morphogen and oncogenic effector protein. Autoprocessing is self-catalyzed by SHhC, the SHh precursor's C-terminal enzymatic domain. A method to screen for small molecule regulators of this process may be of therapeutic value. Here, we describe the development and validation of the first cellular reporter to monitor human SHhC autoprocessing noninvasively in high-throughput compatible plates. The assay couples intracellular SHhC autoprocessing using endogenous cholesterol to the extracellular secretion of the bioluminescent nanoluciferase enzyme. We developed a WT SHhC reporter line for evaluating potential autoprocessing inhibitors by concentration response-dependent suppression of extracellular bioluminescence. Additionally, a conditional mutant SHhC (D46A) reporter line was developed for identifying potential autoprocessing activators by a concentration response-dependent gain of extracellular bioluminescence. The D46A mutation removes a conserved general base that is critical for the activation of the cholesterol substrate. Inducibility of the D46A reporter was established using a synthetic sterol, 2-α carboxy cholestanol, designed to bypass the defect through intramolecular general base catalysis. To facilitate direct nanoluciferase detection in the cell culture media of 1536-well plates, we designed a novel anionic phosphonylated coelenterazine, CLZ-2P, as the nanoluciferase substrate. This new reporter system offers a long-awaited resource for small molecule discovery for cancer and for developmental disorders where SHh ligand biosynthesis is dysregulated.
Collapse
Affiliation(s)
- Daniel A Ciulla
- Chemistry Department, Binghamton University, Binghamton, New York, USA
| | - Patricia Dranchak
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - John L Pezzullo
- State University of New York, College of Environmental Science and Forestry, Syracuse, New York, USA
| | - Rebecca A Mancusi
- Chemistry Department, Binghamton University, Binghamton, New York, USA
| | | | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - José-Luis Giner
- State University of New York, College of Environmental Science and Forestry, Syracuse, New York, USA.
| | - James Inglese
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA; National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | - Brian P Callahan
- Chemistry Department, Binghamton University, Binghamton, New York, USA.
| |
Collapse
|
15
|
Chen J, Ma D, Zeng C, White LV, Zhang H, Teng Y, Lan P. Solasodine suppress MCF7 breast cancer stem-like cells via targeting Hedgehog/Gli1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154448. [PMID: 36116198 DOI: 10.1016/j.phymed.2022.154448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 08/25/2022] [Accepted: 09/05/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Recently, a novel therapy to treat cancer has been to target cancer stem-like cells (CSCs). The aim of this study was to investigate the effect of solasodine, a steroidal alkaloid isolated from Solanum incanum L., on MCF7 CSCs and to understand the compound's underlying mechanism of action. METHOD A tumorsphere formation assay was used to evaluate the effects of solasodine on the proliferation and self-renewal ability of MCF7 CSCs. The level of expression of proteins associated with cancer stemness markers and Hh signaling mediators was determined. The interaction between solasodine and Gli1 was calculated by molecular docking and further demonstrated by cellular thermal shift assay. RESULTS Solasodine significantly decreased the proliferation of MCF7 tumorspheres and showed a stronger cytotoxicity on breast cancer cells with higher levels of Gli1 expression. The results showed that the levels of CD44 and ALDH1 expression were suppressed. Furthermore, expression of CD24 was enhanced by solasodine, via a mechanism that involved dampening Gli1 expression and blocking the nuclear translocation of this protein in MCF7 tumorspheres. Computational studies predicted that solasodine showed a high affinity with the Gli1 zinc finger domain that resulted from hydrogen-bonds to the THR243 and ASP216 amino acids residues. In addition, solasodine specifically bound with Gli1 and enhanced Gli1 protein stability in MCF7 cells. CONCLUSION Here, our findings indicated that solasodine can directly suppresses Hh/Gli1 signaling, and is a novel anticancer candidate that targets CSCs.
Collapse
Affiliation(s)
- Jing Chen
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Da Ma
- College of Packaging engineering, Jinan University, Zhuhai, 519070, China.
| | - Cuicui Zeng
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lorenzo V White
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Huanqing Zhang
- College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yinglai Teng
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Ping Lan
- College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
16
|
Kaur P, Otgonbaatar A, Ramamoorthy A, Chua EHZ, Harmston N, Gruber J, Tolwinski NS. Combining stem cell rejuvenation and senescence targeting to synergistically extend lifespan. Aging (Albany NY) 2022; 14:8270-8291. [PMID: 36287172 PMCID: PMC9648810 DOI: 10.18632/aging.204347] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022]
Abstract
Why biological age is a major risk factor for many of the most important human diseases remains mysterious. We know that as organisms age, stem cell pools are exhausted while senescent cells progressively accumulate. Independently, induction of pluripotency via expression of Yamanaka factors (Oct4, Klf4, Sox2, c-Myc; OKSM) and clearance of senescent cells have each been shown to ameliorate cellular and physiological aspects of aging, suggesting that both processes are drivers of organismal aging. But stem cell exhaustion and cellular senescence likely interact in the etiology and progression of age-dependent diseases because both undermine tissue and organ homeostasis in different if not complementary ways. Here, we combine transient cellular reprogramming (stem cell rejuvenation) with targeted removal of senescent cells to test the hypothesis that simultaneously targeting both cell-fate based aging mechanisms will maximize life and health span benefits. We find that OKSM extends lifespan and show that both interventions protect the intestinal stem cell pool, lower inflammation, activate pro-stem cell signaling pathways, and synergistically improve health and lifespan. Our findings suggest that a combination therapy, simultaneously replacing lost stem cells and removing senescent cells, shows synergistic potential for anti-aging treatments. Our finding that transient expression of both is the most effective suggests that drug-based treatments in non-genetically tractable organisms will likely be the most translatable.
Collapse
Affiliation(s)
- Prameet Kaur
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
| | | | | | | | - Nathan Harmston
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jan Gruber
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
- Department of Biochemistry, NUS, Singapore 117596, Singapore
| | - Nicholas S. Tolwinski
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
17
|
He Y, Chen S, Liu K, Chen Y, Cheng Y, Zeng P, Zhu P, Xie T, Chen S, Zhang H, Cheng J. OsHIPL1, a hedgehog-interacting protein-like 1 protein, increases seed vigour in rice. PLANT BIOTECHNOLOGY JOURNAL 2022; 20:1346-1362. [PMID: 35315188 PMCID: PMC9241377 DOI: 10.1111/pbi.13812] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/19/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
The cultivation of rice varieties with high seed vigour is vital for the direct seeding of rice, and the molecular basis of regulation of seed vigour remains elusive. Here, we cloned a new gene OsHIPL1, which encodes hedgehog-interacting protein-like 1 protein as a causal gene of the major QTL qSV3 for rice seed vigour. OsHIPL1 was mainly localized in the plasma membrane and nucleus. RNA sequencing (RNA-seq) revealed that the ABA-related genes were involved in the OsHIPL1 regulation of seed vigour in rice. The higher levels of endogenous ABA were measured in germinating seeds of OsHIPL1 mutants and NIL-qsv3 line compared to IR26 plants, with two up-regulated ABA biosynthesis genes (OsZEP and OsNCED4) and one down-regulated ABA catabolism gene OsABA8ox3. The expression of abscisic acid-insensitive 3 (OsABI3), OsABI4 and OsABI5 was significantly up-regulated in germinating seeds of OsHIPL1 mutants and NIL-qsv3 line compared to IR26 plants. These results indicate that the regulation of seed vigour of OsHIPL1 may be through modulating endogenous ABA levels and altering OsABIs expression during seed germination in rice. Meanwhile, we found that OsHIPL1 interacted with the aquaporin OsPIP1;1, then affected water uptake to promote rice seed germination. Based on analysis of single-nucleotide polymorphism data of rice accessions, we identified a Hap1 haplotype of OsHIPL1 that was positively correlated with seed germination. Our findings showed novel insights into the molecular mechanism of OsHIPL1 on seed vigour.
Collapse
Affiliation(s)
- Ying He
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Shanshan Chen
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Kexin Liu
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Yongji Chen
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Yanhao Cheng
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Peng Zeng
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Peiwen Zhu
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Ting Xie
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Sunlu Chen
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Hongsheng Zhang
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| | - Jinping Cheng
- State Key Laboratory of Crop Genetics and Germplasm EnhancementJiangsu Collaborative Innovation Center for Modern Crop ProductionJiangsu Province Engineering Research Center of Seed Industry Science and TechnologyCyrus Tang Innovation Center for Seed IndustryNanjing Agricultural UniversityNanjingChina
| |
Collapse
|
18
|
Xu S, Tang C. Cholesterol and Hedgehog Signaling: Mutual Regulation and Beyond. Front Cell Dev Biol 2022; 10:774291. [PMID: 35573688 PMCID: PMC9091300 DOI: 10.3389/fcell.2022.774291] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
The Hedgehog (HH) signaling is one of the key agents that govern the precisely regulated developmental processes of multicellular organisms in vertebrates and invertebrates. The HH pathway in the receiving cell includes Patched1, a twelve-pass transmembrane receptor, and Smoothened, a seven-transmembrane G-protein coupled receptor (GPCR), and the downstream GLI family of three transcriptional factors (GLI1-GLI3). Mutations of HH gene and the main components in HH signaling are also associated with numerous types of diseases. Before secretion, the HH protein undergoes post-translational cholesterol modification to gain full activity, and cholesterol is believed to be essential for proper HH signaling transduction. In addition, results from recent studies show the reciprocal effect that HH signaling functions in cholesterol metabolism as well as in cholesterol homeostasis, which provides feedback to HH pathway. Here, we hope to provide new insights into HH signaling function by discussing the role of cholesterol in HH protein maturation, secretion and HH signaling transduction, and the potential role of HH in regulation of cholesterol as well.
Collapse
|
19
|
Qiu S, Chen J, Kim JT, Zhou Y, Moon JH, Lee SB, Park HJ, Lee HJ. Suppression of Adipogenesis and Fat Accumulation by Vitexin Through Activation of Hedgehog Signaling in 3T3-L1 Adipocytes. J Med Food 2022; 25:313-323. [PMID: 35320011 DOI: 10.1089/jmf.2021.k.0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Many studies have demonstrated that adipogenesis is associated with obesity, and the Hedgehog (Hh) signaling pathway regulates adipogenesis and obesity. Following the screening study of the chemical library evaluating the effect of vitexin on Gli1 transcriptional activity, vitexin was chosen as a candidate for antiadipogenic efficacy. Vitexin significantly reduced lipid accumulation and suppressed C/EBPα (CCAAT/enhancer-binding protein α) and PPARγ (peroxisome proliferator-activated receptor γ) expression, which are known as key adipogenic factors in the early stages of adipogenesis by activating Hh signaling. Furthermore, Hh inhibitor GANT61 reversed the effect of AMP-activated protein kinase (AMPK) activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide), indicating that Hh signaling is an upstream regulator of AMPK in 3T3-L1 cells. Vitexin suppressed adipogenesis by regulating Hh signaling and phosphorylation of AMPK, leading to the inhibition of fat formation. These results suggest that vitexin can be considered a potent dietary agent in alleviating lipid accumulation and obesity.
Collapse
Affiliation(s)
- Shuai Qiu
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Jing Chen
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, China
| | - Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Yimeng Zhou
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Ji Hyun Moon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Seung Beom Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Ho Jin Park
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, South Korea
| |
Collapse
|
20
|
Goodman LD, Bellen HJ. Recent insights into the role of glia and oxidative stress in Alzheimer's disease gained from Drosophila. Curr Opin Neurobiol 2022; 72:32-38. [PMID: 34418791 PMCID: PMC8854453 DOI: 10.1016/j.conb.2021.07.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 02/03/2023]
Abstract
Here, we discuss findings made using Drosophila on Alzheimer's disease (AD) risk and progression. Recent studies have investigated the mechanisms underlying glia-mediated neuroprotection in AD. First, we discuss a novel mechanism of glial lipid droplet formation that occurs in response to elevated reactive oxygen species in neurons. The data suggest that disruptions to this process contribute to AD risk. We further discuss novel mechanistic insights into glia-mediated Aβ42-clearance made using the fly. Finally, we highlight work that provides evidence that the aberrant accumulation of reactive oxygen species in AD may not just be a consequence of disease but contribute to disease progression as well. Cumulatively, the discussed studies highlight recent, relevant discoveries in AD made using Drosophila.
Collapse
Affiliation(s)
- Lindsey D. Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA,CORRESPONDANCE
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
21
|
Maeso‐Díaz R, Dalton GD, Oh S, Du K, Tang L, Chen T, Dutta RK, Hartman JH, Meyer JN, Diehl A. Aging reduces liver resiliency by dysregulating Hedgehog signaling. Aging Cell 2022; 21:e13530. [PMID: 34984806 PMCID: PMC8844109 DOI: 10.1111/acel.13530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/02/2021] [Accepted: 11/28/2021] [Indexed: 11/30/2022] Open
Abstract
Older age is a major risk factor for damage to many tissues, including liver. Aging undermines resiliency and impairs liver regeneration. The mechanisms whereby aging reduces resiliency are poorly understood. Hedgehog is a signaling pathway with critical mitogenic and morphogenic functions during development. Recent studies indicate that Hedgehog regulates metabolic homeostasis in adult liver. The present study evaluates the hypothesis that Hedgehog signaling becomes dysregulated in hepatocytes during aging, resulting in decreased resiliency and therefore, impaired regeneration and enhanced vulnerability to damage. Partial hepatectomy (PH) was performed on young and old wild‐type mice and Smoothened (Smo)‐floxed mice treated with viral vectors to conditionally delete Smo and disrupt Hedgehog signaling specifically in hepatocytes. Changes in signaling were correlated with changes in regenerative responses and compared among groups. Old livers had fewer hepatocytes proliferating after PH. RNA sequencing identified Hedgehog as a top downregulated pathway in old hepatocytes before and after the regenerative challenge. Deleting Smo in young hepatocytes before PH prevented Hedgehog pathway activation after PH and inhibited regeneration. Gene Ontogeny analysis demonstrated that both old and Smo‐deleted young hepatocytes had activation of pathways involved in innate immune responses and suppression of several signaling pathways that control liver growth and metabolism. Hedgehog inhibition promoted telomere shortening and mitochondrial dysfunction in hepatocytes, consequences of aging that promote inflammation and impair tissue growth and metabolic homeostasis. Hedgehog signaling is dysregulated in old hepatocytes. This accelerates aging, resulting in decreased resiliency and therefore, impaired liver regeneration and enhanced vulnerability to damage.
Collapse
Affiliation(s)
- Raquel Maeso‐Díaz
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| | - George D. Dalton
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| | - Sehhoon Oh
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| | - Kuo Du
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| | - Linda Tang
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| | - Tianyi Chen
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| | - Rajesh K. Dutta
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| | - Jessica H. Hartman
- Nicholas School of the Environment Duke University Durham North Carolina USA
| | - Joel N. Meyer
- Nicholas School of the Environment Duke University Durham North Carolina USA
| | - Anna Mae Diehl
- Division of Gastroenterology Department of Medicine Duke University Health System Durham North Carolina USA
| |
Collapse
|
22
|
Kaur P, Chua EHZ, Lim WK, Liu J, Harmston N, Tolwinski NS. Wnt Signaling Rescues Amyloid Beta-Induced Gut Stem Cell Loss. Cells 2022; 11:cells11020281. [PMID: 35053396 PMCID: PMC8774390 DOI: 10.3390/cells11020281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/05/2023] Open
Abstract
Patients with Alzheimer’s disease suffer from a decrease in brain mass and a prevalence of amyloid-β plaques. These plaques are thought to play a role in disease progression, but their exact role is not entirely established. We developed an optogenetic model to induce amyloid-β intracellular oligomerization to model distinct disease etiologies. Here, we examine the effect of Wnt signaling on amyloid in an optogenetic, Drosophila gut stem cell model. We observe that Wnt activation rescues the detrimental effects of amyloid expression and oligomerization. We analyze the gene expression changes downstream of Wnt that contribute to this rescue and find changes in aging related genes, protein misfolding, metabolism, and inflammation. We propose that Wnt expression reduces inflammation through repression of Toll activating factors. We confirm that chronic Toll activation reduces lifespan, but a decrease in the upstream activator Persephone extends it. We propose that the protective effect observed for lithium treatment functions, at least in part, through Wnt activation and the inhibition of inflammation.
Collapse
Affiliation(s)
- Prameet Kaur
- Division of Science, Yale-NUS College, Singapore 138527, Singapore; (P.K.); (E.H.Z.C.); (W.K.L.); (J.L.); (N.H.)
| | - Ellora Hui Zhen Chua
- Division of Science, Yale-NUS College, Singapore 138527, Singapore; (P.K.); (E.H.Z.C.); (W.K.L.); (J.L.); (N.H.)
| | - Wen Kin Lim
- Division of Science, Yale-NUS College, Singapore 138527, Singapore; (P.K.); (E.H.Z.C.); (W.K.L.); (J.L.); (N.H.)
| | - Jiarui Liu
- Division of Science, Yale-NUS College, Singapore 138527, Singapore; (P.K.); (E.H.Z.C.); (W.K.L.); (J.L.); (N.H.)
| | - Nathan Harmston
- Division of Science, Yale-NUS College, Singapore 138527, Singapore; (P.K.); (E.H.Z.C.); (W.K.L.); (J.L.); (N.H.)
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Nicholas S. Tolwinski
- Division of Science, Yale-NUS College, Singapore 138527, Singapore; (P.K.); (E.H.Z.C.); (W.K.L.); (J.L.); (N.H.)
- Correspondence: ; Tel.: +65-66013092
| |
Collapse
|
23
|
Sarkar A, Nazir A. Carrying Excess Baggage Can Slowdown Life: Protein Clearance Machineries That Go Awry During Aging and the Relevance of Maintaining Them. Mol Neurobiol 2021; 59:821-840. [PMID: 34792731 DOI: 10.1007/s12035-021-02640-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023]
Abstract
Cellular homeostasis is maintained by rapid and systematic cleansing of aberrant and aggregated proteins within cells. Neurodegenerative diseases (NDs) especially Parkinson's and Alzheimer's disease are known to be associated with multiple factors, most important being impaired clearance of aggregates, resulting in the accumulation of specific aggregated protein in the brain. Protein quality control (PQC) of proteostasis network comprises proteolytic machineries and chaperones along with their regulators to ensure precise operation and maintenance of proteostasis. Such regulatory factors coordinate among each other multiple functional aspects related to proteins, including their synthesis, folding, transport, and degradation. During aging due to inevitable endogenous and external stresses, sustaining a proteome balance is a challenging task. Such stresses decline the capacity of the proteostasis network compromising the proteome integrity, affecting the fundamental physiological processes including reproductive fitness of the organism. This review focuses on highlighting proteome-wide changes during aging and the strategies for proteostasis improvements. The possibility of augmenting the proteostasis network either via genetic or pharmacological interventions may be a promising strategy towards delaying age-associated pathological consequences due to proteome disbalance, thus promoting healthy aging and prolonged longevity.
Collapse
Affiliation(s)
- Arunabh Sarkar
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, 226031, India
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, 226031, India.
| |
Collapse
|
24
|
Babagana M, Oh KS, Chakraborty S, Pacholewska A, Aqdas M, Sung MH. Hedgehog dysregulation contributes to tissue-specific inflammaging of resident macrophages. Aging (Albany NY) 2021; 13:19207-19229. [PMID: 34390567 PMCID: PMC8386529 DOI: 10.18632/aging.203422] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/03/2021] [Indexed: 12/31/2022]
Abstract
Age-associated low-grade sterile inflammation, commonly referred to as inflammaging, is a recognized hallmark of aging, which contributes to many age-related diseases. While tissue-resident macrophages are innate immune cells that secrete many types of inflammatory cytokines in response to various stimuli, it is not clear whether they have a role in driving inflammaging. Here we characterized the transcriptional changes associated with physiological aging in mouse resident macrophage populations across different tissues and sexes. Although the age-related transcriptomic signatures of resident macrophages were strikingly tissue-specific, the differentially expressed genes were collectively enriched for those with important innate immune functions such as antigen presentation, cytokine production, and cell adhesion. The brain-resident microglia had the most wide-ranging age-related alterations, with compromised expression of tissue-specific genes and relatively exaggerated responses to endotoxin stimulation. Despite the tissue-specific patterns of aging transcriptomes, components of the hedgehog (Hh) signaling pathway were decreased in aged macrophages across multiple tissues. In vivo suppression of Hh signaling in young animals increased the expression of pro-inflammatory cytokines, while in vitro activation of Hh signaling in old macrophages, in turn, suppressed the expression of these inflammatory cytokines. This suggests that hedgehog signaling could be a potential intervention axis for mitigating age-associated inflammation and related diseases. Overall, our data represent a resourceful catalog of tissue-specific and sex-specific transcriptomic changes in resident macrophages of peritoneum, liver, and brain, during physiological aging.
Collapse
Affiliation(s)
- Mahamat Babagana
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kyu-Seon Oh
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Sayantan Chakraborty
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Alicja Pacholewska
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.,Present Address: Institute for Translational Epigenetics, University Hospital Cologne, Cologne, Germany
| | - Mohammad Aqdas
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myong-Hee Sung
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
25
|
Elder MK, Erdjument-Bromage H, Oliveira MM, Mamcarz M, Neubert TA, Klann E. Age-dependent shift in the de novo proteome accompanies pathogenesis in an Alzheimer's disease mouse model. Commun Biol 2021; 4:823. [PMID: 34193971 PMCID: PMC8245541 DOI: 10.1038/s42003-021-02324-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder associated with memory loss, but the AD-associated neuropathological changes begin years before memory impairments. Investigation of the early molecular abnormalities in AD might offer innovative opportunities to target memory impairment prior to onset. Decreased protein synthesis plays a fundamental role in AD, yet the consequences of this dysregulation for cellular function remain unknown. We hypothesize that alterations in the de novo proteome drive early metabolic alterations in the hippocampus that persist throughout AD progression. Using a combinatorial amino acid tagging approach to selectively label and enrich newly synthesized proteins, we found that the de novo proteome is disturbed in young APP/PS1 mice prior to symptom onset, affecting the synthesis of multiple components of the synaptic, lysosomal, and mitochondrial pathways. Furthermore, the synthesis of large clusters of ribosomal subunits were affected throughout development. Our data suggest that large-scale changes in protein synthesis could underlie cellular dysfunction in AD.
Collapse
Affiliation(s)
- Megan K Elder
- Center for Neural Science, New York University, New York, NY, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | | | - Maggie Mamcarz
- Center for Neural Science, New York University, New York, NY, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, USA.
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
26
|
Nita A, Abraham SP, Krejci P, Bosakova M. Oncogenic FGFR Fusions Produce Centrosome and Cilia Defects by Ectopic Signaling. Cells 2021; 10:1445. [PMID: 34207779 PMCID: PMC8227969 DOI: 10.3390/cells10061445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
A single primary cilium projects from most vertebrate cells to guide cell fate decisions. A growing list of signaling molecules is found to function through cilia and control ciliogenesis, including the fibroblast growth factor receptors (FGFR). Aberrant FGFR activity produces abnormal cilia with deregulated signaling, which contributes to pathogenesis of the FGFR-mediated genetic disorders. FGFR lesions are also found in cancer, raising a possibility of cilia involvement in the neoplastic transformation and tumor progression. Here, we focus on FGFR gene fusions, and discuss the possible mechanisms by which they function as oncogenic drivers. We show that a substantial portion of the FGFR fusion partners are proteins associated with the centrosome cycle, including organization of the mitotic spindle and ciliogenesis. The functions of centrosome proteins are often lost with the gene fusion, leading to haploinsufficiency that induces cilia loss and deregulated cell division. We speculate that this complements the ectopic FGFR activity and drives the FGFR fusion cancers.
Collapse
Affiliation(s)
- Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Sara P. Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| |
Collapse
|
27
|
Shi H, Sun F, Yang T, Peng M, Wang M, Zhang Y, Wang Y, Dong C, Yan Z, Si G, Wang W, Li Y. Construction of a ceRNA immunoregulatory network related to the development of vascular dementia through a weighted gene coexpression network analysis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:858. [PMID: 34164492 PMCID: PMC8184445 DOI: 10.21037/atm-21-1717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background To date, vascular dementia (VaD) lacks effective treatment in clinical practice. There is also growing evidence that VaD may be closely related to the immune response. The development of high-throughput technology, and the recently discovered group of new mediators called competitive endogenous RNAs (ceRNA), provides a unique opportunity to study the immunomodulation of VaD. Methods In this study, we used gene expression profiles in the Gene Expression Omnibus (GEO) database to obtain immune-related gene coexpression modules through a weighted gene coexpression network analysis (WGCNA) and gene enrichment analysis. We extracted and merged long non-coding RNA (lncRNA) and microRNA (miRNA) expressions from the GEO database and mapped them with related databases. Subsequently, we used Cytoscape to construct a lncRNA-miRNA-mRNA network, and then we performed an enrichment analysis on the mRNAs in the network to determine their regulatory function. Subsequently, we used an ImmuCellAI immune infiltration analysis and constructed a ceRNA sub-network of related immune cells. Finally, we conducted a gene set enrichment analysis (GSEA) to determine the potential regulatory pathways of the key factors. Results As a result, we identified the blue module as the key module of immunity and constructed the related CeRNA network. Immune infiltration analysis showed that natural killer T (NKT) cells may be the key immune cells of VaD. Using a Pearson correlation analysis, we identified that B4GALT1, PPP1R3B, MICB, HHAT, DSC2, DNA2, SCARA3, and lncRNA NEAT1 may be the key factors of VaD. Our subsequent GSEA analysis showed that lncRNA NEAT1 may be regulated by NK cells and toll-like receptors. Conclusions Our research provides new therapeutic targets for vascular dementia from the immunological perspective for the first time, including B4GALT1, PPP1R3B, MICB, HHAT, DSC2, DNA2, SCARA3, and lncRNA NEAT1, and our research hopes to provide new treatment options for VaD.
Collapse
Affiliation(s)
- Hongshuo Shi
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengshan Sun
- Department of Encephalopathy, Jinan Traditional Chinese Medicine Hospital, Jinan, China
| | - Tiantian Yang
- Department of Traditional Chinese Medicine, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Min Peng
- Department of Traditional Chinese Medicine, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Min Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiwen Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengda Dong
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhaojun Yan
- Department of Psychosomatic Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guomin Si
- Department of Traditional Chinese Medicine, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wenbo Wang
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Orthopaedics, The 8th Clinical College (Weihai Central Hospital), Qingdao University, Qingdao, China
| | - Yujie Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
28
|
Miller HA, Dean ES, Pletcher SD, Leiser SF. Cell non-autonomous regulation of health and longevity. eLife 2020; 9:62659. [PMID: 33300870 PMCID: PMC7728442 DOI: 10.7554/elife.62659] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/28/2022] Open
Abstract
As the demographics of the modern world skew older, understanding and mitigating the effects of aging is increasingly important within biomedical research. Recent studies in model organisms demonstrate that the aging process is frequently modified by an organism’s ability to perceive and respond to changes in its environment. Many well-studied pathways that influence aging involve sensory cells, frequently neurons, that signal to peripheral tissues and promote survival during the presence of stress. Importantly, this activation of stress response pathways is often sufficient to improve health and longevity even in the absence of stress. Here, we review the current landscape of research highlighting the importance of cell non-autonomous signaling in modulating aging from C. elegans to mammals. We also discuss emerging concepts including retrograde signaling, approaches to mapping these networks, and development of potential therapeutics.
Collapse
Affiliation(s)
- Hillary A Miller
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, United States
| | - Elizabeth S Dean
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States
| | - Scott D Pletcher
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States
| | - Scott F Leiser
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| |
Collapse
|
29
|
Frey B, AlOkda A, Jackson MP, Riguet N, Duce JA, Lashuel HA. Monitoring alpha-synuclein oligomerization and aggregation using bimolecular fluorescence complementation assays: What you see is not always what you get. J Neurochem 2020; 157:872-888. [PMID: 32772367 PMCID: PMC8246987 DOI: 10.1111/jnc.15147] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022]
Abstract
Bimolecular fluorescence complementation (BiFC) was introduced a decade ago as a method to monitor alpha‐synuclein (α‐syn) oligomerization in intact cells. Since then, several α‐syn BiFC cellular assays and animal models have been developed based on the assumption that an increase in the fluorescent signal correlates with increased α‐syn oligomerization or aggregation. Despite the increasing use of these assays and models in mechanistic studies, target validation and drug screening, there have been no reports that (1) validate the extent to which the BiFC fluorescent signal correlates with α‐syn oligomerization at the biochemical level; (2) provide a structural characterization of the oligomers and aggregates formed by the BiFC. To address this knowledge gap, we first analysed the expression level and oligomerization properties of the individual constituents of α‐syn‐Venus, one of the most commonly used BiFC systems, in HEK‐293 & SH‐SY5Y cells from three different laboratories using multiple biochemical approaches and techniques. Next, we investigated the biochemical and aggregation properties of α‐syn upon co‐expression of both BiFC fragments. Our results show that (1) the C‐terminal‐Venus fused to α‐syn (α‐syn‐Vc) is present in much lower abundance than its counterpart with N‐terminal‐Venus fused to α‐syn (Vn‐α‐syn); (2) Vn‐α‐syn exhibits a high propensity to form oligomers and higher‐order aggregates; and (3) the expression of either or both fragments does not result in the formation of α‐syn fibrils or cellular inclusions. Furthermore, our results suggest that only a small fraction of Vn‐α‐syn is involved in the formation of the fluorescent BiFC complex and that some of the fluorescent signal may arise from the association or entrapment of α‐syn‐Vc in Vn‐α‐syn aggregates. The fact that the N‐terminal fragment exists predominantly in an aggregated state also indicates that one must exercise caution when using this system to investigate α‐syn oligomerization in cells or in vivo. Altogether, our results suggest that cellular and animal models of oligomerization, aggregation and cell‐to‐cell transmission based on the α‐syn BiFC systems should be thoroughly characterized at the biochemical level to ensure that they reproduce the process of interest and measure what they are intended to measure. ![]()
Collapse
Affiliation(s)
- Bryan Frey
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Abdelrahman AlOkda
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthew P Jackson
- The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Nathan Riguet
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - James A Duce
- The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
30
|
Zarkali A, McColgan P, Ryten M, Reynolds RH, Leyland LA, Lees AJ, Rees G, Weil RS. Dementia risk in Parkinson's disease is associated with interhemispheric connectivity loss and determined by regional gene expression. Neuroimage Clin 2020; 28:102470. [PMID: 33395965 PMCID: PMC7581968 DOI: 10.1016/j.nicl.2020.102470] [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: 06/14/2020] [Revised: 09/08/2020] [Accepted: 10/11/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's dementia is a common and devastating part of Parkinson's disease. Whilst timing and severity vary, dementia in Parkinson's is often preceded by visual dysfunction. White matter changes, representing axonal loss, occur early in the disease process. Clarifying which white matter connections are affected in Parkinson's with visual dysfunction and why specific connections are vulnerable will provide important mechanistic insights. Here, we use diffusion tractography in 100 Parkinson's patients (33 low visual performers) and 34 controls to identify patterns of connectivity loss in Parkinson's with visual dysfunction. We examine the relationship between regional transcription and connectivity loss, using the Allen Institute for Brain Science transcriptome atlas. We show that interhemispheric connections are preferentially affected in Parkinson's low visual performers. Interhemispheric connection loss was associated with downweighted genes related to the smoothened signalling pathway (enriched in glutamatergic neurons) and upweighted metabolic genes. Risk genes for Parkinson's but not Alzheimer's or Dementia with Lewy bodies were over-represented in upweighted genes associated with interhemispheric connection loss. Our findings suggest selective vulnerability in Parkinson's patients at highest risk of dementia (those with visual dysfunction), where differences in gene expression and metabolic dysfunction, affecting longer connections with higher metabolic burden, drive connectivity loss.
Collapse
Affiliation(s)
- Angeliki Zarkali
- Dementia Research Centre, University College London, 8-11 Queen Square, London WC1N 3AR, UK.
| | - Peter McColgan
- Huntington's Disease Centre, University College London, Russell Square House, London WC1B 5EH, UK
| | - Mina Ryten
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK; Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, 10-12 Russell Square House, London WC1B 5EH, UK
| | - Regina H Reynolds
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK; Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, 10-12 Russell Square House, London WC1B 5EH, UK
| | - Louise-Ann Leyland
- Dementia Research Centre, University College London, 8-11 Queen Square, London WC1N 3AR, UK
| | - Andrew J Lees
- Reta Lila Weston Institute of Neurological Studies, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Geraint Rees
- Institute of Cognitive Neuroscience, University College London, 17-19 Queen Square, London WC1N 3AR, UK; Wellcome Centre for Human Neuroimaging, University College London, 12 Queen Square, London WC1N 3AR, UK
| | - Rimona S Weil
- Dementia Research Centre, University College London, 8-11 Queen Square, London WC1N 3AR, UK; Wellcome Centre for Human Neuroimaging, University College London, 12 Queen Square, London WC1N 3AR, UK; Movement Disorders Consortium, University College London, London WC1N 3BG, UK
| |
Collapse
|
31
|
Johnson AA, Shokhirev MN, Wyss-Coray T, Lehallier B. Systematic review and analysis of human proteomics aging studies unveils a novel proteomic aging clock and identifies key processes that change with age. Ageing Res Rev 2020; 60:101070. [PMID: 32311500 DOI: 10.1016/j.arr.2020.101070] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
The development of clinical interventions that significantly improve human healthspan requires robust markers of biological age as well as thoughtful therapeutic targets. To promote these goals, we performed a systematic review and analysis of human aging and proteomics studies. The systematic review includes 36 different proteomics analyses, each of which identified proteins that significantly changed with age. We discovered 1,128 proteins that had been reported by at least two or more analyses and 32 proteins that had been reported by five or more analyses. Each of these 32 proteins has known connections relevant to aging and age-related disease. GDF15, for example, extends both lifespan and healthspan when overexpressed in mice and is additionally required for the anti-diabetic drug metformin to exert beneficial effects on body weight and energy balance. Bioinformatic enrichment analyses of our 1,128 commonly identified proteins heavily implicated processes relevant to inflammation, the extracellular matrix, and gene regulation. We additionally propose a novel proteomic aging clock comprised of proteins that were reported to change with age in plasma in three or more different studies. Using a large patient cohort comprised of 3,301 subjects (aged 18-76 years), we demonstrate that this clock is able to accurately predict human age.
Collapse
|