1
|
Astacio E, DiAngelo JR. The regulation of triglyceride storage by Acsx4 and Acsx5 in Drosophila fat tissue. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001430. [PMID: 39958912 PMCID: PMC11830124 DOI: 10.17912/micropub.biology.001430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/18/2025] [Accepted: 02/01/2025] [Indexed: 02/18/2025]
Abstract
The production of energy is one of the most fundamental requirements for organismal survival. Decreasing expression of Drosophila 9G8 , an mRNA splicing protein, specifically in adipose tissue results in triglyceride accumulation. Decreasing 9G8 in adipose also results in upregulation of the acyl-CoA synthetases Acsx4 and Acsx5 ; however, the functions of these genes in regulating lipid metabolism is not fully understood. Here, we decreased Acsx4 and Acsx5 in fly adipose tissue and this resulted in high triglycerides. This suggests that these genes regulate lipid breakdown, and their upregulation is perhaps compensating for the triglyceride accumulation observed when 9G8 levels are decreased.
Collapse
|
2
|
Ye X, Li Y, González-Lamuño D, Pei Z, Moser AB, Smith KD, Watkins PA. Role of ACSBG1 in Brain Lipid Metabolism and X-Linked Adrenoleukodystrophy Pathogenesis: Insights from a Knockout Mouse Model. Cells 2024; 13:1687. [PMID: 39451204 PMCID: PMC11506745 DOI: 10.3390/cells13201687] [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: 08/26/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
"Bubblegum" acyl-CoA synthetase (ACSBG1) is a pivotal player in lipid metabolism during mouse brain development, facilitating the activation of long-chain fatty acids (LCFA) and their incorporation into lipid species that are crucial for brain function. ACSBG1 converts LCFA into acyl-CoA derivatives, supporting vital metabolic processes. Fruit fly mutants lacking ACSBG1 exhibited neurodegeneration and had elevated levels of very long-chain fatty acids (VLCFA), characteristics of human X-linked adrenoleukodystrophy (XALD). To explore ACSBG1's function and potential as a therapeutic target in XALD, we created an ACSBG1 knockout (Acsbg1-/-) mouse and examined the effects on brain FA metabolism during development. Phenotypically, Acsbg1-/- mice resembled wild type (w.t.) mice. ACSBG1 expression was found mainly in tissue affected pathologically in XALD, namely the brain, adrenal gland and testis. ACSBG1 depletion did not significantly reduce the total ACS enzyme activity in these tissue types. In adult mouse brain, ACSBG1 expression was highest in the cerebellum; the low levels detected during the first week of life dramatically increased thereafter. Unexpectedly, lower, rather than higher, saturated VLCFA levels were found in cerebella from Acsbg1-/- vs. w.t. mice, especially after one week of age. Developmental changes in monounsaturated ω9 FA and polyunsaturated ω3 FA levels also differed between w.t. and Acsbg1-/- mice. ACSBG1 deficiency impacted the developmental expression of several cerebellar FA metabolism enzymes, including those required for the synthesis of ω3 polyunsaturated FA, precursors of bioactive signaling molecules like eicosanoids and docosanoids. These changes in membrane lipid FA composition likely affect membrane fluidity and may thus influence the body's response to inflammation. We conclude that, despite compelling circumstantial evidence, it is unlikely that ACSBG1 directly contributes to the pathology of XALD, decreasing its potential as a therapeutic target. Instead, the effects of ACSBG1 knockout on processes regulated by eicosanoids and/or docosanoids should be further investigated.
Collapse
Affiliation(s)
- Xiaoli Ye
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yuanyuan Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Genetic Medicine and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Domingo González-Lamuño
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhengtong Pei
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ann B. Moser
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kirby D. Smith
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Genetic Medicine and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul A. Watkins
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
3
|
Manor J, Jangam SV, Chung HL, Bhagwat P, Andrews J, Chester H, Kondo S, Srivastav S, Botas J, Moser AB, Huguenin SM, Wangler MF. Genetic analysis of the X-linked Adrenoleukodystrophy ABCD1 gene in Drosophila uncovers a role in Peroxisomal dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614586. [PMID: 39386423 PMCID: PMC11463603 DOI: 10.1101/2024.09.23.614586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a progressive neurodegenerative disorder caused by a loss-of-function (LOF) mutation in the ATP-binding cassette subfamily D member 1 (ABCD1) gene, leading to the accumulation of very long-chain fatty acids (VLCFAs). This disorder exhibits striking heterogeneity; some male patients develop an early childhood neuroinflammatory demyelination disorder, while other patients, including adult males and most affected female carriers, experience a chronic progressive myelopathy. Adrenocortical failure is observed in almost all male patients, with age of onset varying sometimes being the first diagnostic finding. The gene underlying this spectrum of disease encodes an ATP-binding cassette (ABC) transporter that localizes to peroxisomes and facilitates VLCFA transport. X-ALD is considered a single peroxisomal component defect and does not play a direct role in peroxisome assembly. Drosophila models of other peroxisomal genes have provided mechanistic insight into some of the neurodegenerative mechanisms with reduced lifespan, retinal degeneration, and VLCFA accumulation. Here, we perform a genetic analysis of the fly ABCD1 ortholog Abcd1 (CG2316). Knockdown or deficiency of Abcd1 leads to VLCFA accumulation, salivary gland defects, locomotor impairment and retinal lipid abnormalities. Interestingly, there is also evidence of reduced peroxisomal numbers. Flies overexpressing the human cDNA for ABCD1 display a wing crumpling phenotype characteristic of the pex2 loss-of-function. Surprisingly, overexpression of human ABCD1 appears to inhibit or overwhelm peroxisomal biogenesis to levels similar to null mutations in fly pex2, pex16 and pex3. Drosophila Abcd1 is therefore implicated in peroxisomal number, and overexpression of the human ABCD1 gene acts a potent inhibitor of peroxisomal biogenesis in flies.
Collapse
Affiliation(s)
- Joshua Manor
- Metabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Sharayu V Jangam
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Hyung-lok Chung
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
- Department of Neurology, Weill Cornell Medical College, New York, NY, USA
| | - Pranjali Bhagwat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Jonathan Andrews
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Hillary Chester
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Shu Kondo
- Tokyo University of Science, Faculty of Advanced Engineering, Department of Biological Science and Technology, Tokyo, Japan
| | - Saurabh Srivastav
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Ann B. Moser
- Hugo W Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Suzette M. Huguenin
- Hugo W Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
4
|
Daenzer JMI, Druss JJ, Fridovich-Keil JL. Restoring galactose metabolism without restoring GALT rescues both compromised survival in larvae and an adult climbing deficit in a GALT-null D. Melanogaster model of classic galactosemia. J Inherit Metab Dis 2024; 47:991-1000. [PMID: 38960603 PMCID: PMC11563841 DOI: 10.1002/jimd.12774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
Classic galactosemia (CG) is an autosomal recessive disorder that results from profound deficiency of galactose-1-phosphate uridylyltransferase (GALT), the middle enzyme in the highly conserved Leloir pathway of galactose metabolism. That galactose metabolism is disrupted in patients with CG, and in GALT-null microbial, cell culture, and animal models of CG, has been known for many years. However, whether the long-term developmental complications of CG result from disrupted galactose metabolism alone, or from loss of some independent moonlighting function of GALT, in addition to disrupted galactose metabolism, has been posed but never resolved. Here, we addressed this question using a GALT-null Drosophila melanogaster model of CG engineered to express uridine diphosphate (UDP)-glucose/galactose pyrophosphorylase (UGGP), a plant enzyme that effectively bypasses GALT in the Leloir pathway by converting substrates uridine triphosphate (UTP) plus galactose-1-phosphate (gal-1P) into products UDP-galactose plus pyrophosphate (PPi). While GALT and UGGP share one substrate (gal-1P) and one product (UDP-galactose), they are structurally and evolutionarily unrelated enzymes. It is therefore extremely unlikely that they would also share a moonlighting function. We found that GALT-null flies expressing UGGP showed not only partial rescue of metabolic abnormalities and acute larval sensitivity to dietary galactose, as expected, but also full rescue of an adult motor deficit otherwise seen in this model. By extension, these results may offer insights to the underlying bases of at least some acute and long-term complications experienced by patients with CG.
Collapse
Affiliation(s)
| | - Jared J. Druss
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia USA
| | | |
Collapse
|
5
|
Ye X, Li Y, González-Lamuño D, Pei Z, Moser AB, Smith KD, Watkins PA. Role of ACSBG1 in brain lipid metabolism and X-linked adrenoleukodystrophy pathogenesis: Insights from a knockout mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599741. [PMID: 38948805 PMCID: PMC11212999 DOI: 10.1101/2024.06.19.599741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The "bubblegum" acyl-CoA synthetase (ACSBG1) is a pivotal player in lipid metabolism during the development of the mouse brain, facilitating the activation of long-chain fatty acids (LCFAs) and their integration into essential lipid species crucial for brain function. Through its enzymatic activity, ACSBG1 converts LCFAs into acyl-CoA derivatives, supporting vital processes like membrane formation, myelination, and energy production. Its regulatory role significantly influences neuronal growth, synaptic plasticity, and overall brain development, highlighting its importance in maintaining lipid homeostasis and proper brain function. Originally discovered in the fruit fly brain, ACSBG1 attracted attention for its potential implication in X-linked adrenoleukodystrophy (XALD) pathogenesis. Studies using Drosophila melanogaster lacking the ACSBG1 homolog, bubblegum, revealed adult neurodegeneration with elevated levels of very long-chain fatty acids (VLCFA). To explore ACSBG1's role in fatty acid (FA) metabolism and its relevance to XALD, we created an ACSBG1 knockout (Acsbg1-/-) mouse model and examined its impact on lipid metabolism during mouse brain development. Phenotypically, Acsbg1-/- mice resembled wild type (w.t.) mice. Despite its primary expression in tissues affected by XALD, brain, adrenal gland and testis, ACSBG1 depletion did not significantly reduce total ACS enzyme activity in these tissues when using LCFA or VLCFA as substrates. However, analysis unveiled intriguing developmental and compositional changes in FA levels associated with ACSBG1 deficiency. In the adult mouse brain, ACSBG1 expression peaked in the cerebellum, with lower levels observed in other brain regions. Developmentally, ACSBG1 expression in the cerebellum was initially low during the first week of life but increased dramatically thereafter. Cerebellar FA levels were assessed in both w.t. and Acsbg1-/- mouse brains throughout development, revealing notable differences. While saturated VLCFA levels were typically high in XALD tissues and in fruit flies lacking ACSBG1, cerebella from Acsbg1-/- mice displayed lower saturated VLCFA levels, especially after about 8 days of age. Additionally, monounsaturated ω9 FA levels exhibited a similar trend as saturated VLCFA, while ω3 polyunsaturated FA levels were elevated in Acsbg1-/- mice. Further analysis of specific FA levels provided additional insights into potential roles for ACSBG1. Notably, the decreased VLCFA levels in Acsbg1-/- mice primarily stemmed from changes in C24:0 and C26:0, while reduced ω9 FA levels were mainly observed in C18:1 and C24:1. ACSBG1 depletion had minimal effects on saturated long-chain FA or ω6 polyunsaturated FA levels but led to significant increases in specific ω3 FA, such as C20:5 and C22:5. Moreover, the impact of ACSBG1 deficiency on the developmental expression of several cerebellar FA metabolism enzymes, including those required for synthesis of ω3 polyunsaturated FA, was assessed; these FA can potentially be converted into bioactive signaling molecules like eicosanoids and docosanoids. In conclusion, despite compelling circumstantial evidence, it is unlikely that ACSBG1 directly contributes to the pathology of XALD. Instead, the effects of ACSBG1 knockout on processes regulated by eicosanoids and/or docosanoids should be further investigated.
Collapse
Affiliation(s)
- Xiaoli Ye
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Present address: School of Life Sciences, Southwest University, Chongqing, China
| | - Yuanyuan Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
- Department of Genetic Medicine and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Present address: Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467
| | - Domingo González-Lamuño
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Present address: Pediatra. Unidad de Nefrología y Metabolismo Infantil, Hospital U. Marqués de Valdecilla. Santander. España
| | - Zhengtong Pei
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ann B. Moser
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Kirby D. Smith
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
- Department of Genetic Medicine and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Paul A. Watkins
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
6
|
Maruoka N, Makino T, Urabe J. RNA-seq analysis to identify genes related to resting egg production of panarctic Daphnia pulex. BMC Genomics 2023; 24:262. [PMID: 37198540 PMCID: PMC10190107 DOI: 10.1186/s12864-023-09369-3] [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: 02/17/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The genus Daphnia switches its reproductive mode from subitaneous egg production to resting egg production in response to environmental stimuli. Although this life history trait is essential for surviving unsuitable environments, the molecular mechanism of resting egg production is little understood. In this study, we examined genes related to induction of resting egg production using two genotypes of panarctic Daphnia pulex, the JPN1 and JPN2 lineages, which differ genetically in the frequency of resting egg production. We reared these genotypes under high and low food levels. At the high food level, individuals of both genotypes continually produced subitaneous eggs, whereas at the low food level, only the JPN2 genotype produced resting eggs. Then, we performed RNA-seq analysis on specimens of three instars, including before and after egg production. RESULTS These results showed that expressed genes differed significantly between individuals grown under high and low food levels and among individuals of different instars and genotypes. Among these differentially expressed genes (DEGs), we found 16 that changed expression level before resting egg production. Some of these genes showed high-level expression only before resting egg production and one gene was an ortholog of bubblegum (bgm), which is reportedly up-regulated before diapause in bumblebees. According to gene ontology (GO) enrichment analysis, one GO term annotated as long-chain fatty acid biosynthetic process was enriched among these 16 genes. In addition, GO terms related to glycometabolism were enriched among down-regulated genes of individuals holding resting eggs, compared to those before resting egg production. CONCLUSIONS We found candidate genes highly expressed only before resting egg production. Although functions of candidate genes found in this study have not been reported previously in Daphnia, catabolism of long-chain fatty acids and metabolism of glycerates are related to diapause in other organisms. Thus, it is highly probable that candidate genes identified in this study are related to the molecular mechanism regulating resting egg production in Daphnia.
Collapse
Affiliation(s)
- Natsumi Maruoka
- Graduate School of Life sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, 022-795-6686, Miyagi, Japan.
- Center for Bioscience Research and Education, Utsunomiya University, 350 Mine-machi, Utsunomiya, 321-8505, 028-649-5129, Tochigi, Japan.
| | - Takashi Makino
- Graduate School of Life sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, 022-795-6686, Miyagi, Japan
| | - Jotaro Urabe
- Graduate School of Life sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, 022-795-6686, Miyagi, Japan
| |
Collapse
|
7
|
Bujdoso R, Smith A, Fleck O, Spiropoulos J, Andréoletti O, Thackray AM. Prion disease modelled in Drosophila. Cell Tissue Res 2023; 392:47-62. [PMID: 35092497 PMCID: PMC10113284 DOI: 10.1007/s00441-022-03586-0] [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: 08/27/2021] [Accepted: 01/17/2022] [Indexed: 11/02/2022]
Abstract
Prion diseases are fatal neurodegenerative conditions of humans and various vertebrate species that are transmissible between individuals of the same or different species. A novel infectious moiety referred to as a prion is considered responsible for transmission of these conditions. Prion replication is believed to be the cause of the neurotoxicity that arises during prion disease pathogenesis. The prion hypothesis predicts that the transmissible prion agent consists of PrPSc, which is comprised of aggregated misfolded conformers of the normal host protein PrPC. It is important to understand the biology of transmissible prions and to identify genetic modifiers of prion-induced neurotoxicity. This information will underpin the development of therapeutic and control strategies for human and animal prion diseases. The most reliable method to detect prion infectivity is by in vivo transmission in a suitable experimental host, which to date have been mammalian species. Current prion bioassays are slow, cumbersome and relatively insensitive to low titres of prion infectivity, and do not lend themselves to rapid genetic analysis of prion disease. Here, we provide an overview of our novel studies that have led to the establishment of Drosophila melanogaster, a genetically well-defined invertebrate host, as a sensitive, versatile and economically viable animal model for the detection of mammalian prion infectivity and genetic modifiers of prion-induced toxicity.
Collapse
Affiliation(s)
- Raymond Bujdoso
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK.
| | - Andrew Smith
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK
| | - Oliver Fleck
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK
| | - John Spiropoulos
- Pathology Department, Animal and Plant Health Agency (APHA), Weybridge, Woodham Lane, New Haw, Surrey, KT15 3NB, Addlestone, UK
| | - Olivier Andréoletti
- UMR INRA ENVT 1225-Hôtes-Agents Pathogènes, Ecole Nationale Vétérinaire de Toulouse, 23 Chemin des Capelles, 31076, Toulouse, France
| | - Alana M Thackray
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK.
| |
Collapse
|
8
|
Suh GSB, Yu K, Kim YJ, Oh Y, Park JJ. History of Drosophila neurogenetic research in South Korea. J Neurogenet 2022:1-7. [PMID: 36165786 DOI: 10.1080/01677063.2022.2115040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Neurogenetic research using the Drosophila model has immensely expanded around the world. Likewise, scientists in South Korea have leveraged the advantages of Drosophila genetic tools to understand various neurobiological processes. In this special issue, we will overview the history of Drosophila neurogenetic research in South Korea that led to significant discoveries and notably implications. We will describe how Drosophila system was first introduced to elevate neural developmental studies in 1990s. Establishing Drosophila-related resources has been a key venture, which led to the generation of over 100,000 mutant lines and the launch of the K-Gut initiative with Korea Drosophila Research Center (KDRC). These resources have supported the pioneer studies in modeling human disease and understanding genes and neural circuits that regulate animal behavior and physiology.
Collapse
Affiliation(s)
- Greg S B Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kweon Yu
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Metabolism and Neurophysiology Research Group, Daejeon, Republic of Korea
| | - Young-Joon Kim
- Department of Biological Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yangkyun Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Joong-Jean Park
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Manor J, Chung H, Bhagwat PK, Wangler MF. ABCD1 and X-linked adrenoleukodystrophy: A disease with a markedly variable phenotype showing conserved neurobiology in animal models. J Neurosci Res 2021; 99:3170-3181. [PMID: 34716609 PMCID: PMC9665428 DOI: 10.1002/jnr.24953] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a phenotypically heterogeneous disorder involving defective peroxisomal β-oxidation of very long-chain fatty acids (VLCFAs), due to mutation in the ABCD1 gene. X-ALD is the most common peroxisomal inborn error of metabolism and confers a high degree of morbidity and mortality. Remarkably, a subset of patients exhibit a cerebral form with inflammatory invasion of the central nervous system and extensive demyelination, while in others only dying-back axonopathy or even isolated adrenal insufficiency is seen, without genotype-phenotype correlation. X-ALD's biochemical signature is marked elevation of VLCFAs in blood, a finding that has been utilized for massive newborn screening for early diagnosis. Investigational gene therapy approaches hold promises for improved outcomes. However, the pathophysiological mechanisms of the disease remain poorly understood, limiting investigation of targeted therapeutic options. Animal models for the disease recapitulate the biochemical signature of VLCFA accumulation and demonstrate mitochondrially generated reactive oxygen species, oxidative damage, increased glial death, and axonal damage. Most strikingly, however, cerebral invasion of leukocytes and demyelination were not observed in any animal model for X-ALD, reflecting upon pathological processes that are yet to be discovered. This review summarizes the current disease models in animals, the lessons learned from these models, and the gaps that remained to be filled in order to assist in therapeutic investigations for ALD.
Collapse
Affiliation(s)
- Joshua Manor
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | - Hyunglok Chung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Pranjali K. Bhagwat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Michael F. Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
10
|
The 50th anniversary of the Konopka and Benzer 1971 paper in PNAS: "Clock Mutants of Drosophila melanogaster". Proc Natl Acad Sci U S A 2021; 118:2110171118. [PMID: 34507977 DOI: 10.1073/pnas.2110171118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 11/18/2022] Open
|
11
|
Montoro R, Heine VM, Kemp S, Engelen M. Evolution of adrenoleukodystrophy model systems. J Inherit Metab Dis 2021; 44:544-553. [PMID: 33373044 PMCID: PMC8248356 DOI: 10.1002/jimd.12357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 01/09/2023]
Abstract
X-linked adrenoleukodystrophy (ALD) is a neurometabolic disorder affecting the adrenal glands, testes, spinal cord and brain. The disease is caused by mutations in the ABCD1 gene resulting in a defect in peroxisomal degradation of very long-chain fatty acids and their accumulation in plasma and tissues. Males with ALD have a near 100% life-time risk to develop myelopathy. The life-time prevalence to develop progressive cerebral white matter lesions (known as cerebral ALD) is about 60%. Adrenal insufficiency occurs in about 80% of male patients. In adulthood, 80% of women with ALD also develop myelopathy, but adrenal insufficiency or cerebral ALD are very rare. The complex clinical presentation and the absence of a genotype-phenotype correlation are complicating our understanding of the disease. In an attempt to understand the pathophysiology of ALD various model systems have been developed. While these model systems share the basic genetics and biochemistry of ALD they fail to fully recapitulate the complex neurodegenerative etiology of ALD. Each model system recapitulates certain aspects of the disorder. This exposes the complexity of ALD and therefore the challenge to create a comprehensive model system to fully understand ALD. In this review, we provide an overview of the different ALD modeling strategies from single-celled to multicellular organisms and from in vitro to in vivo approaches, and introduce how emerging iPSC-derived technologies could improve the understanding of this highly complex disorder.
Collapse
Affiliation(s)
- Roberto Montoro
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Vivi M. Heine
- Department of Child and Youth Psychiatry, Amsterdam UMC, Amsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Stephan Kemp
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marc Engelen
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
12
|
Billey E, Magneschi L, Leterme S, Bedhomme M, Andres-Robin A, Poulet L, Michaud M, Finazzi G, Dumas R, Crouzy S, Laueffer F, Fourage L, Rébeillé F, Amato A, Collin S, Jouhet J, Maréchal E. Characterization of the Bubblegum acyl-CoA synthetase of Microchloropsis gaditana. PLANT PHYSIOLOGY 2021; 185:815-835. [PMID: 33793914 PMCID: PMC8133546 DOI: 10.1093/plphys/kiaa110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/15/2020] [Indexed: 05/15/2023]
Abstract
The metabolic pathways of glycerolipids are well described in cells containing chloroplasts limited by a two-membrane envelope but not in cells containing plastids limited by four membranes, including heterokonts. Fatty acids (FAs) produced in the plastid, palmitic and palmitoleic acids (16:0 and 16:1), are used in the cytosol for the synthesis of glycerolipids via various routes, requiring multiple acyl-Coenzyme A (CoA) synthetases (ACS). Here, we characterized an ACS of the Bubblegum subfamily in the photosynthetic eukaryote Microchloropsis gaditana, an oleaginous heterokont used for the production of lipids for multiple applications. Genome engineering with TALE-N allowed the generation of MgACSBG point mutations, but no knockout was obtained. Point mutations triggered an overall decrease of 16:1 in lipids, a specific increase of unsaturated 18-carbon acyls in phosphatidylcholine and decrease of 20-carbon acyls in the betaine lipid diacylglyceryl-trimethyl-homoserine. The profile of acyl-CoAs highlighted a decrease in 16:1-CoA and 18:3-CoA. Structural modeling supported that mutations affect accessibility of FA to the MgACSBG reaction site. Expression in yeast defective in acyl-CoA biosynthesis further confirmed that point mutations affect ACSBG activity. Altogether, this study supports a critical role of heterokont MgACSBG in the production of 16:1-CoA and 18:3-CoA. In M. gaditana mutants, the excess saturated and monounsaturated FAs were diverted to triacylglycerol, thus suggesting strategies to improve the oil content in this microalga.
Collapse
Affiliation(s)
- Elodie Billey
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
- Total Raffinage-Chimie, Tour Coupole, 2 Place Jean Millier, 92078 Paris La Défense, France
| | - Leonardo Magneschi
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Sébastien Leterme
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Mariette Bedhomme
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
- Total Raffinage-Chimie, Tour Coupole, 2 Place Jean Millier, 92078 Paris La Défense, France
| | - Amélie Andres-Robin
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Laurent Poulet
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Morgane Michaud
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Giovanni Finazzi
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Renaud Dumas
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Serge Crouzy
- Laboratoire de Chimie et Biologie des Métaux, Unité mixte de Recherche 5249 CNRS–CEA–Univ. Grenoble Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Frédéric Laueffer
- Total Raffinage-Chimie, Tour Coupole, 2 Place Jean Millier, 92078 Paris La Défense, France
| | - Laurent Fourage
- Total Raffinage-Chimie, Tour Coupole, 2 Place Jean Millier, 92078 Paris La Défense, France
| | - Fabrice Rébeillé
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Alberto Amato
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Séverine Collin
- Total Raffinage-Chimie, Tour Coupole, 2 Place Jean Millier, 92078 Paris La Défense, France
| | - Juliette Jouhet
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| | - Eric Maréchal
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de Recherche 5168 CNRS–CEA–INRA–Univ. Grenoble-Alpes, IRIG, CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
| |
Collapse
|
13
|
Genetic Underpinnings of Host Manipulation by Ophiocordyceps as Revealed by Comparative Transcriptomics. G3-GENES GENOMES GENETICS 2020; 10:2275-2296. [PMID: 32354705 PMCID: PMC7341126 DOI: 10.1534/g3.120.401290] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ant-infecting Ophiocordyceps fungi are globally distributed, host manipulating, specialist parasites that drive aberrant behaviors in infected ants, at a lethal cost to the host. An apparent increase in activity and wandering behaviors precedes a final summiting and biting behavior onto vegetation, which positions the manipulated ant in a site beneficial for fungal growth and transmission. We investigated the genetic underpinnings of host manipulation by: (i) producing a high-quality hybrid assembly and annotation of the Ophiocordyceps camponoti-floridani genome, (ii) conducting laboratory infections coupled with RNAseq of O. camponoti-floridani and its host, Camponotus floridanus, and (iii) comparing these data to RNAseq data of Ophiocordyceps kimflemingiae and Camponotus castaneus as a powerful method to identify gene expression patterns that suggest shared behavioral manipulation mechanisms across Ophiocordyceps-ant species interactions. We propose differentially expressed genes tied to ant neurobiology, odor response, circadian rhythms, and foraging behavior may result by activity of putative fungal effectors such as enterotoxins, aflatrem, and mechanisms disrupting feeding behaviors in the ant.
Collapse
|
14
|
The Drosophila melanogaster as Genetic Model System to Dissect the Mechanisms of Disease that Lead to Neurodegeneration in Adrenoleukodystrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1299:145-159. [PMID: 33417213 DOI: 10.1007/978-3-030-60204-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Drosophila melanogaster is the most successful genetic model organism to study different human disease with a recent increased popularity to study neurological disorders. Drosophila melanogaster has a complex yet well-defined brain with defined anatomical regions with specific functions. The neuronal network in the adult brain has a structural organization highly similar to human neurons, but in a brain that is much more amenable for complex analyses. The availability of sophisticated genetic tools to study neurons permits to examine neuronal functions at the single cell level in the whole brain by confocal imaging, which does not require sections. Thus, Drosophila has been used to successfully study many neurological disorders such as Parkinson's disease and has been recently adopted to understand the complex networks leading to neurological disorders with metabolic origins such as Leigh disease and X-linked adrenoleukodystrophy (X-ALD).In this review, we will describe the genetic tools available to study neuronal structures and functions and also illustrate some limitations of the system. Finally, we will report the experimental efforts that in the past 10 years have established Drosophila melanogaster as an excellent model organism to study neurodegenerative disorders focusing on X-ALD.
Collapse
|
15
|
Ugrankar R, Bowerman J, Hariri H, Chandra M, Chen K, Bossanyi MF, Datta S, Rogers S, Eckert KM, Vale G, Victoria A, Fresquez J, McDonald JG, Jean S, Collins BM, Henne WM. Drosophila Snazarus Regulates a Lipid Droplet Population at Plasma Membrane-Droplet Contacts in Adipocytes. Dev Cell 2019; 50:557-572.e5. [PMID: 31422916 PMCID: PMC7446143 DOI: 10.1016/j.devcel.2019.07.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 04/22/2019] [Accepted: 07/17/2019] [Indexed: 01/22/2023]
Abstract
Adipocytes store nutrients as lipid droplets (LDs), but how they organize their LD stores to balance lipid uptake, storage, and mobilization remains poorly understood. Here, using Drosophila fat body (FB) adipocytes, we characterize spatially distinct LD populations that are maintained by different lipid pools. We identify peripheral LDs (pLDs) that make close contact with the plasma membrane (PM) and are maintained by lipophorin-dependent lipid trafficking. pLDs are distinct from larger cytoplasmic medial LDs (mLDs), which are maintained by FASN1-dependent de novo lipogenesis. We find that sorting nexin CG1514 or Snazarus (Snz) associates with pLDs and regulates LD homeostasis at ER-PM contact sites. Loss of SNZ perturbs pLD organization, whereas Snz over-expression drives LD expansion, triacylglyceride production, starvation resistance, and lifespan extension through a DESAT1-dependent pathway. We propose that Drosophila adipocytes maintain spatially distinct LD populations and identify Snz as a regulator of LD organization and inter-organelle crosstalk.
Collapse
Affiliation(s)
- Rupali Ugrankar
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jade Bowerman
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hanaa Hariri
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Mintu Chandra
- Institute for Molecular Bioscience, the University of Queensland, St. Lucia, QLD 4072, Australia
| | - Kevin Chen
- Institute for Molecular Bioscience, the University of Queensland, St. Lucia, QLD 4072, Australia
| | - Marie-France Bossanyi
- Department of Anatomy and Cell Biology, University of Sherbrooke, 2500 Boulevard de l'Universite, Sherbrooke, QC J1K 2R1, Canada
| | - Sanchari Datta
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Sean Rogers
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Kaitlyn M Eckert
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gonçalo Vale
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alexia Victoria
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | - Jeffrey G McDonald
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Steve Jean
- Department of Anatomy and Cell Biology, University of Sherbrooke, 2500 Boulevard de l'Universite, Sherbrooke, QC J1K 2R1, Canada
| | - Brett M Collins
- Institute for Molecular Bioscience, the University of Queensland, St. Lucia, QLD 4072, Australia
| | - W Mike Henne
- Department of Cell Biology, UT Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
16
|
Thackray AM, Andréoletti O, Bujdoso R. Mammalian prion propagation in PrP transgenic Drosophila. Brain 2019; 141:2700-2710. [PMID: 29985975 PMCID: PMC6113635 DOI: 10.1093/brain/awy183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/24/2018] [Indexed: 12/22/2022] Open
Abstract
Mammalian prions propagate by template-directed misfolding and aggregation of normal cellular prion related protein PrPC as it converts into disease-associated conformers collectively referred to as PrPSc. Mammalian species may be permissive for prion disease because these hosts have co-evolved specific co-factors that assist PrPC conformational change and prion propagation. We have tested this hypothesis by examining whether faithful prion propagation occurs in the normally PrPC-null invertebrate host Drosophila melanogaster. Ovine PrP transgenic Drosophila exposed at the larval stage to ovine scrapie showed a progressive accumulation of transmissible prions in adult flies. Strikingly, the biological properties of distinct ovine prion strains were maintained during their propagation in Drosophila. Our observations show that the co-factors necessary for strain-specific prion propagation are not unique to mammalian species. Our studies establish Drosophila as a novel host for the study of transmissible mammalian prions.
Collapse
Affiliation(s)
- Alana M Thackray
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, Cambridge, CB3 OES, UK
| | - Olivier Andréoletti
- UMR INRA ENVT 1225 -Hôtes-Agents Pathogènes, Ecole Nationale Vétérinaire de Toulouse, 23 Chemin des Capelles, Toulouse, France
| | - Raymond Bujdoso
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, Cambridge, CB3 OES, UK
| |
Collapse
|
17
|
Baião GC, Schneider DI, Miller WJ, Klasson L. The effect of Wolbachia on gene expression in Drosophila paulistorum and its implications for symbiont-induced host speciation. BMC Genomics 2019; 20:465. [PMID: 31174466 PMCID: PMC6555960 DOI: 10.1186/s12864-019-5816-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 05/21/2019] [Indexed: 11/17/2022] Open
Abstract
Background The Neotropical fruit fly Drosophila paulistorum (Diptera: Drosophilidae) is a species complex in statu nascendi comprising six reproductively isolated semispecies, each harboring mutualistic Wolbachia strains. Although wild type flies of each semispecies are isolated from the others by both pre- and postmating incompatibilities, mating between semispecies and successful offspring development can be achieved once flies are treated with antibiotics to reduce Wolbachia titer. Here we use RNA-seq to study the impact of Wolbachia on D. paulistorum and investigate the hypothesis that the symbiont may play a role in host speciation. For that goal, we analyze samples of heads and abdomens of both sexes of the Amazonian, Centro American and Orinocan semispecies of D. paulistorum. Results We identify between 175 and 1192 differentially expressed genes associated with a variety of biological processes that respond either globally or according to tissue, sex or condition in the three semispecies. Some of the functions associated with differentially expressed genes are known to be affected by Wolbachia in other species, such as metabolism and immunity, whereas others represent putative novel phenotypes involving muscular functions, pheromone signaling, and visual perception. Conclusions Our results show that Wolbachia affect a large number of biological functions in D. paulistorum, particularly when present in high titer. We suggest that the significant metabolic impact of the infection on the host may cause several of the other putative and observed phenotypes. We also speculate that the observed differential expression of genes associated with chemical communication and reproduction may be associated with the emergence of pre- and postmating barriers between semispecies, which supports a role for Wolbachia in the speciation of D. paulistorum. Electronic supplementary material The online version of this article (10.1186/s12864-019-5816-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guilherme C Baião
- Molecular evolution, Department of Cell and Molecular Biology, Uppsala University, Husargatan 3, 751 24, Uppsala, Sweden
| | - Daniela I Schneider
- Lab Genome Dynamics, Deparment Cell & Developmental Biology, Center of Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria.,Present address: Department of Epidemiology of Microbial Diseases, Yale University, 60 College Street, New Haven, CT, 06510, USA
| | - Wolfgang J Miller
- Lab Genome Dynamics, Deparment Cell & Developmental Biology, Center of Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Lisa Klasson
- Molecular evolution, Department of Cell and Molecular Biology, Uppsala University, Husargatan 3, 751 24, Uppsala, Sweden.
| |
Collapse
|
18
|
Triacylglycerol Metabolism in Drosophila melanogaster. Genetics 2019; 210:1163-1184. [PMID: 30523167 DOI: 10.1534/genetics.118.301583] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022] Open
Abstract
Triacylglycerol (TAG) is the most important caloric source with respect to energy homeostasis in animals. In addition to its evolutionarily conserved importance as an energy source, TAG turnover is crucial to the metabolism of structural and signaling lipids. These neutral lipids are also key players in development and disease. Here, we review the metabolism of TAG in the Drosophila model system. Recently, the fruit fly has attracted renewed attention in research due to the unique experimental approaches it affords in studying the tissue-autonomous and interorgan regulation of lipid metabolism in vivo Following an overview of the systemic control of fly body fat stores, we will cover lipid anabolic, enzymatic, and regulatory processes, which begin with the dietary lipid breakdown and de novo lipogenesis that results in lipid droplet storage. Next, we focus on lipolytic processes, which mobilize storage TAG to make it metabolically accessible as either an energy source or as a building block for biosynthesis of other lipid classes. Since the buildup and breakdown of fat involves various organs, we highlight avenues of lipid transport, which are at the heart of functional integration of organismic lipid metabolism. Finally, we draw attention to some "missing links" in basic neutral lipid metabolism and conclude with a perspective on how fly research can be exploited to study functional metabolic roles of diverse lipids.
Collapse
|
19
|
Monnier V, Llorens JV, Navarro JA. Impact of Drosophila Models in the Study and Treatment of Friedreich's Ataxia. Int J Mol Sci 2018; 19:E1989. [PMID: 29986523 PMCID: PMC6073496 DOI: 10.3390/ijms19071989] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
Drosophila melanogaster has been for over a century the model of choice of several neurobiologists to decipher the formation and development of the nervous system as well as to mirror the pathophysiological conditions of many human neurodegenerative diseases. The rare disease Friedreich’s ataxia (FRDA) is not an exception. Since the isolation of the responsible gene more than two decades ago, the analysis of the fly orthologue has proven to be an excellent avenue to understand the development and progression of the disease, to unravel pivotal mechanisms underpinning the pathology and to identify genes and molecules that might well be either disease biomarkers or promising targets for therapeutic interventions. In this review, we aim to summarize the collection of findings provided by the Drosophila models but also to go one step beyond and propose the implications of these discoveries for the study and cure of this disorder. We will present the physiological, cellular and molecular phenotypes described in the fly, highlighting those that have given insight into the pathology and we will show how the ability of Drosophila to perform genetic and pharmacological screens has provided valuable information that is not easily within reach of other cellular or mammalian models.
Collapse
Affiliation(s)
- Véronique Monnier
- Unité de Biologie Fonctionnelle et Adaptative (BFA), Sorbonne Paris Cité, Université Paris Diderot, UMR8251 CNRS, 75013 Paris, France.
| | - Jose Vicente Llorens
- Department of Genetics, University of Valencia, Campus of Burjassot, 96100 Valencia, Spain.
| | - Juan Antonio Navarro
- Lehrstuhl für Entwicklungsbiologie, Universität Regensburg, 93040 Regensburg, Germany.
| |
Collapse
|
20
|
Gordon HB, Valdez L, Letsou A. Etiology and treatment of adrenoleukodystrophy: new insights from Drosophila. Dis Model Mech 2018; 11:dmm031286. [PMID: 29739804 PMCID: PMC6031365 DOI: 10.1242/dmm.031286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/30/2018] [Indexed: 12/25/2022] Open
Abstract
Adrenoleukodystrophy (ALD) is a fatal progressive neurodegenerative disorder affecting brain white matter. The most common form of ALD is X-linked (X-ALD) and results from mutation of the ABCD1-encoded very-long-chain fatty acid (VLCFA) transporter. X-ALD is clinically heterogeneous, with the cerebral form being the most severe. Diagnosed in boys usually between the ages of 4 and 8 years, cerebral X-ALD symptoms progress rapidly (in as little as 2 years) through declines in cognition, learning and behavior, to paralysis and ultimately to a vegetative state and death. Currently, there are no good treatments for X-ALD. Here, we exploit the Drosophila bubblegum (bgm) double bubble (dbb) model of neurometabolic disease to expand diagnostic power and therapeutic potential for ALD. We show that loss of the Drosophila long-/very-long-chain acyl-CoA synthetase genes bgm and/or dbb is indistinguishable from loss of the Drosophila ABC transporter gene ABCD Shared loss-of-function phenotypes for synthetase and transporter mutants point to a lipid metabolic pathway association with ALD-like neurodegenerative disease in Drosophila; a pathway association that has yet to be established in humans. We also show that manipulation of environment increases the severity of neurodegeneration in bgm and dbb mutant flies, adding even further to a suite of new candidate ALD disease-causing genes and pathways in humans. Finally, we show that it is a lack of lipid metabolic pathway product and not (as commonly thought) an accumulation of pathway precursor that is causative of neurometabolic disease: addition of medium-chain fatty acids to the diet of bgm or dbb mutant flies prevents the onset of neurodegeneration. Taken together, our data provide new foundations both for diagnosing ALD and for designing effective, mechanism-based treatment protocols.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Hannah B Gordon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Lourdes Valdez
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Anthea Letsou
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
21
|
Lopes-Marques M, Machado AM, Ruivo R, Fonseca E, Carvalho E, Castro LFC. Expansion, retention and loss in the Acyl-CoA synthetase "Bubblegum" (Acsbg) gene family in vertebrate history. Gene 2018; 664:111-118. [PMID: 29694909 DOI: 10.1016/j.gene.2018.04.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 10/17/2022]
Abstract
Fatty acids (FAs) constitute a considerable fraction of all lipid molecules with a fundamental role in numerous physiological processes. In animals, the majority of complex lipid molecules are derived from the transformation of FAs through several biochemical pathways. Yet, for FAs to enroll in these pathways they require an activation step. FA activation is catalyzed by the rate limiting action of Acyl-CoA synthases. Several Acyl-CoA enzyme families have been previously described and classified according to the chain length of FAs they process. Here, we address the evolutionary history of the ACSBG gene family which activates, FAs with >16 carbons. Currently, two different ACSBG gene families, ACSBG1 and ACSBG2, are recognized in vertebrates. We provide evidence that a wider and unequal ACSBG gene repertoire is present in vertebrate lineages. We identify a novel ACSBG-like gene lineage which occurs specifically in amphibians, ray finned fishes, coelacanths and cartilaginous fishes named ACSBG3. Also, we show that the ACSBG2 gene lineage duplicated in the Theria ancestor. Our findings, thus offer a far richer understanding on FA activation in vertebrates and provide key insights into the relevance of comparative and functional analysis to perceive physiological differences, namely those related with lipid metabolic pathways.
Collapse
Affiliation(s)
- Mónica Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U. Porto), Matosinhos, Portugal.
| | - André M Machado
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U. Porto), Matosinhos, Portugal
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U. Porto), Matosinhos, Portugal
| | - Elza Fonseca
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U. Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U. Porto), Porto, Portugal
| | - Estela Carvalho
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U. Porto), Matosinhos, Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U. Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), Department of Biology, University of Porto (U. Porto), Porto, Portugal.
| |
Collapse
|
22
|
Thimgan MS, Kress N, Lisse J, Fiebelman C, Hilderbrand T. The acyl-CoA Synthetase, pudgy, Promotes Sleep and Is Required for the Homeostatic Response to Sleep Deprivation. Front Endocrinol (Lausanne) 2018; 9:464. [PMID: 30186232 PMCID: PMC6110854 DOI: 10.3389/fendo.2018.00464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The regulation of sleep and the response to sleep deprivation rely on multiple biochemical pathways. A critical connection is the link between sleep and metabolism. Metabolic changes can disrupt sleep, and conversely decreased sleep can alter the metabolic environment. There is building evidence that lipid metabolism, in particular, is a critical part of mounting the homeostatic response to sleep deprivation. We have evaluated an acyl-CoA synthetase, pudgy (pdgy), for its role in sleep and response to sleep deprivation. When pdgy transcript levels are decreased through transposable element disruption of the gene, mutant flies showed lower total sleep times and increased sleep fragmentation at night compared to genetic controls. Consistent with disrupted sleep, mutant flies had a decreased lifespan compared to controls. pdgy disrupted fatty acid handling as pdgy mutants showed increased sensitivity to starvation and exhibited lower fat stores. Moreover, the response to sleep deprivation is reduced when compared to a control flies. When we decreased the transcript levels for pdgy using RNAi, the response to sleep deprivation was decreased compared to background controls. In addition, when the pdgy transcription is rescued throughout the fly, the response to sleep deprivation is restored. These data demonstrate that the regulation and function of acyl-CoA synthetase plays a critical role in regulating sleep and the response to sleep deprivation. Endocrine and metabolic signals that alter transcript levels of pdgy impact sleep regulation or interfere with the homeostatic response to sleep deprivation.
Collapse
Affiliation(s)
- Matthew S. Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
- *Correspondence: Matthew S. Thimgan
| | - Natalie Kress
- Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Josh Lisse
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Courtney Fiebelman
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Thomas Hilderbrand
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| |
Collapse
|
23
|
The lipolysis pathway sustains normal and transformed stem cells in adult Drosophila. Nature 2016; 538:109-113. [PMID: 27680705 DOI: 10.1038/nature19788] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/18/2016] [Indexed: 01/18/2023]
Abstract
Cancer stem cells (CSCs) may be responsible for tumour dormancy, relapse and the eventual death of most cancer patients. In addition, these cells are usually resistant to cytotoxic conditions. However, very little is known about the biology behind this resistance to therapeutics. Here we investigated stem-cell death in the digestive system of adult Drosophila melanogaster. We found that knockdown of the coat protein complex I (COPI)-Arf79F (also known as Arf1) complex selectively killed normal and transformed stem cells through necrosis, by attenuating the lipolysis pathway, but spared differentiated cells. The dying stem cells were engulfed by neighbouring differentiated cells through a draper-myoblast city-Rac1-basket (also known as JNK)-dependent autophagy pathway. Furthermore, Arf1 inhibitors reduced CSCs in human cancer cell lines. Thus, normal or cancer stem cells may rely primarily on lipid reserves for energy, in such a way that blocking lipolysis starves them to death. This finding may lead to new therapies that could help to eliminate CSCs in human cancers.
Collapse
|
24
|
Huo X, Wu B, Feng M, Han B, Fang Y, Hao Y, Meng L, Wubie AJ, Fan P, Hu H, Qi Y, Li J. Proteomic Analysis Reveals the Molecular Underpinnings of Mandibular Gland Development and Lipid Metabolism in Two Lines of Honeybees (Apis mellifera ligustica). J Proteome Res 2016; 15:3342-57. [DOI: 10.1021/acs.jproteome.6b00526] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Xinmei Huo
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Bin Wu
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Mao Feng
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Bin Han
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Yu Fang
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Yue Hao
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Lifeng Meng
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Abebe Jenberie Wubie
- Department
of Animal production and Technology, College of Agriculture and Environmental
Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Pei Fan
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Han Hu
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Yuping Qi
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| | - Jianke Li
- Institute
of Apicultural Research/Key Laboratory of Pollinating Insect Biology,
Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing 100093, China
| |
Collapse
|
25
|
Becker N, Kucharski R, Rössler W, Maleszka R. Age-dependent transcriptional and epigenomic responses to light exposure in the honey bee brain. FEBS Open Bio 2016; 6:622-39. [PMID: 27398303 PMCID: PMC4932443 DOI: 10.1002/2211-5463.12084] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 01/21/2023] Open
Abstract
Light is a powerful environmental stimulus of special importance in social honey bees that undergo a behavioral transition from in-hive to outdoor foraging duties. Our previous work has shown that light exposure induces structural neuronal plasticity in the mushroom bodies (MBs), a brain center implicated in processing inputs from sensory modalities. Here, we extended these analyses to the molecular level to unravel light-induced transcriptomic and epigenomic changes in the honey bee brain. We have compared gene expression in brain compartments of 1- and 7-day-old light-exposed honey bees with age-matched dark-kept individuals. We have found a number of differentially expressed genes (DEGs), both novel and conserved, including several genes with reported roles in neuronal plasticity. Most of the DEGs show age-related changes in the amplitude of light-induced expression and are likely to be both developmentally and environmentally regulated. Some of the DEGs are either known to be methylated or are implicated in epigenetic processes suggesting that responses to light exposure are at least partly regulated at the epigenome level. Consistent with this idea light alters the DNA methylation pattern of bgm, one of the DEGs affected by light exposure, and the expression of microRNA miR-932. This confirms the usefulness of our approach to identify candidate genes for neuronal plasticity and provides evidence for the role of epigenetic processes in driving the molecular responses to visual stimulation.
Collapse
Affiliation(s)
- Nils Becker
- Behavioral Physiology and Sociobiology Biozentrum University of Würzburg Germany
| | - Robert Kucharski
- Research School of Biology The Australian National University Acton Australia
| | - Wolfgang Rössler
- Behavioral Physiology and Sociobiology Biozentrum University of Würzburg Germany
| | - Ryszard Maleszka
- Research School of Biology The Australian National University Acton Australia
| |
Collapse
|
26
|
Sivachenko A, Gordon HB, Kimball SS, Gavin EJ, Bonkowsky JL, Letsou A. Neurodegeneration in a Drosophila model of adrenoleukodystrophy: the roles of the Bubblegum and Double bubble acyl-CoA synthetases. Dis Model Mech 2016; 9:377-87. [PMID: 26893370 PMCID: PMC4852500 DOI: 10.1242/dmm.022244] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 02/17/2016] [Indexed: 12/21/2022] Open
Abstract
Debilitating neurodegenerative conditions with metabolic origins affect millions of individuals worldwide. Still, for most of these neurometabolic disorders there are neither cures nor disease-modifying therapies, and novel animal models are needed for elucidation of disease pathology and identification of potential therapeutic agents. To date, metabolic neurodegenerative disease has been modeled in animals with only limited success, in part because existing models constitute analyses of single mutants and have thus overlooked potential redundancy within metabolic gene pathways associated with disease. Here, we present the first analysis of a very-long-chain acyl-CoA synthetase (ACS) double mutant. We show that the Drosophila bubblegum(bgm) and double bubble(dbb) genes have overlapping functions, and that the consequences of double knockout of both bubblegum and double bubble in the fly brain are profound, affecting behavior and brain morphology, and providing the best paradigm to date for an animal model of adrenoleukodystrophy (ALD), a fatal childhood neurodegenerative disease associated with the accumulation of very-long-chain fatty acids. Using this more fully penetrant model of disease to interrogate brain morphology at the level of electron microscopy, we show that dysregulation of fatty acid metabolism via disruption of ACS function in vivois causal of neurodegenerative pathologies that are evident in both neuronal cells and their supporting cell populations, and leads ultimately to lytic cell death in affected areas of the brain. Finally, in an extension of our model system to the study of human disease, we describe our identification of an individual with leukodystrophy who harbors a rare mutation in SLC27a6(encoding a very-long-chain ACS), a human homolog of bgm and dbb.
Collapse
Affiliation(s)
- Anna Sivachenko
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Hannah B Gordon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Suzanne S Kimball
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Erin J Gavin
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah, Salt Lake City, UT 84112, USA
| | - Anthea Letsou
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
27
|
Bousquet F, Chauvel I, Flaven-Pouchon J, Farine JP, Ferveur JF. Dietary rescue of altered metabolism gene reveals unexpected Drosophila mating cues. J Lipid Res 2016; 57:443-50. [PMID: 26759364 DOI: 10.1194/jlr.m064683] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Indexed: 11/20/2022] Open
Abstract
To develop and reproduce, animals need long-chain MUFAs and PUFAs. Although some unsaturated FAs (UFAs) can be synthesized by the organism, others must be provided by the diet. The gene, desat1, involved in Drosophila melanogaster UFA metabolism, is necessary for both larval development and for adult sex pheromone communication. We first characterized desat1 expression in larval tissues. Then, we found that larvae in which desat1 expression was knocked down throughout development died during the larval stages when raised on standard food. By contrast pure MUFAs or PUFAs, but not saturated FAs, added to the larval diet rescued animals to adulthood with the best effect being obtained with oleic acid (C18:1). Male and female mating behavior and fertility were affected very differently by preimaginal UFA-rich diet. Adult diet also strongly influenced several aspects of reproduction: flies raised on a C18:1-rich diet showed increased mating performance compared with flies raised on standard adult diet. Therefore, both larval and adult desat1 expression control sex-specific mating signals. A similar nutrigenetics approach may be useful in other metabolic mutants to uncover cryptic effects otherwise masked by severe developmental defects.
Collapse
Affiliation(s)
- François Bousquet
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS, UMR 1324 INRA, University of Burgundy, F-21000 Dijon, France
| | - Isabelle Chauvel
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS, UMR 1324 INRA, University of Burgundy, F-21000 Dijon, France
| | - Justin Flaven-Pouchon
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS, UMR 1324 INRA, University of Burgundy, F-21000 Dijon, France Centro Interdisciplinario de Neurociencia de Valparaiso, University of Valparaiso, Valparaiso, Chile
| | - Jean-Pierre Farine
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS, UMR 1324 INRA, University of Burgundy, F-21000 Dijon, France
| | - Jean-François Ferveur
- Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS, UMR 1324 INRA, University of Burgundy, F-21000 Dijon, France
| |
Collapse
|
28
|
|
29
|
Xu Z, Tito AJ, Rui YN, Zhang S. Studying polyglutamine diseases in Drosophila. Exp Neurol 2015; 274:25-41. [PMID: 26257024 DOI: 10.1016/j.expneurol.2015.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 08/02/2015] [Accepted: 08/03/2015] [Indexed: 12/16/2022]
Abstract
Polyglutamine (polyQ) diseases are a family of dominantly transmitted neurodegenerative disorders caused by an abnormal expansion of CAG trinucleotide repeats in the protein-coding regions of the respective disease-causing genes. Despite their simple genetic basis, the etiology of these diseases is far from clear. Over the past two decades, Drosophila has proven to be successful in modeling this family of neurodegenerative disorders, including the faithful recapitulation of pathological features such as polyQ length-dependent formation of protein aggregates and progressive neuronal degeneration. Additionally, it has been valuable in probing the pathogenic mechanisms, in identifying and evaluating disease modifiers, and in helping elucidate the normal functions of disease-causing genes. Knowledge learned from this simple invertebrate organism has had a large impact on our understanding of these devastating brain diseases.
Collapse
Affiliation(s)
- Zhen Xu
- The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Medical School at Houston, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Health Science Center at Houston (UTHealth), 1825 Pressler Street, Houston, TX 77030, United States
| | - Antonio Joel Tito
- The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, United States; Programs in Human and Molecular Genetics and Neuroscience, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Graduate School of Biomedical Sciences, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Medical School at Houston, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Health Science Center at Houston (UTHealth), 1825 Pressler Street, Houston, TX 77030, United States
| | - Yan-Ning Rui
- The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Medical School at Houston, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Health Science Center at Houston (UTHealth), 1825 Pressler Street, Houston, TX 77030, United States
| | - Sheng Zhang
- The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, United States; Department of Neurobiology and Anatomy, 1825 Pressler Street, Houston, TX 77030, United States; Programs in Human and Molecular Genetics and Neuroscience, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Graduate School of Biomedical Sciences, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Medical School at Houston, 1825 Pressler Street, Houston, TX 77030, United States; The University of Texas Health Science Center at Houston (UTHealth), 1825 Pressler Street, Houston, TX 77030, United States.
| |
Collapse
|
30
|
Reiff T, Jacobson J, Cognigni P, Antonello Z, Ballesta E, Tan KJ, Yew JY, Dominguez M, Miguel-Aliaga I. Endocrine remodelling of the adult intestine sustains reproduction in Drosophila. eLife 2015. [PMID: 26216039 PMCID: PMC4515472 DOI: 10.7554/elife.06930] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The production of offspring is energetically costly and relies on incompletely understood mechanisms that generate a positive energy balance. In mothers of many species, changes in key energy-associated internal organs are common yet poorly characterised functionally and mechanistically. In this study, we show that, in adult Drosophila females, the midgut is dramatically remodelled to enhance reproductive output. In contrast to extant models, organ remodelling does not occur in response to increased nutrient intake and/or offspring demands, but rather precedes them. With spatially and temporally directed manipulations, we identify juvenile hormone (JH) as an anticipatory endocrine signal released after mating. Acting through intestinal bHLH-PAS domain proteins Methoprene-tolerant (Met) and Germ cell-expressed (Gce), JH signals directly to intestinal progenitors to yield a larger organ, and adjusts gene expression and sterol regulatory element-binding protein (SREBP) activity in enterocytes to support increased lipid metabolism. Our findings identify a metabolically significant paradigm of adult somatic organ remodelling linking hormonal signals, epithelial plasticity, and reproductive output.
Collapse
Affiliation(s)
- Tobias Reiff
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas, Universidad Miguel Hernández, Alicante, Spain
| | - Jake Jacobson
- MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Paola Cognigni
- MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Zeus Antonello
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas, Universidad Miguel Hernández, Alicante, Spain
| | - Esther Ballesta
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas, Universidad Miguel Hernández, Alicante, Spain
| | - Kah Junn Tan
- Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Joanne Y Yew
- Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Maria Dominguez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas, Universidad Miguel Hernández, Alicante, Spain
| | - Irene Miguel-Aliaga
- MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| |
Collapse
|
31
|
Thimgan MS, Seugnet L, Turk J, Shaw PJ. Identification of genes associated with resilience/vulnerability to sleep deprivation and starvation in Drosophila. Sleep 2015; 38:801-14. [PMID: 25409104 DOI: 10.5665/sleep.4680] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/10/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND STUDY OBJECTIVES Flies mutant for the canonical clock protein cycle (cyc(01)) exhibit a sleep rebound that is ∼10 times larger than wild-type flies and die after only 10 h of sleep deprivation. Surprisingly, when starved, cyc(01) mutants can remain awake for 28 h without demonstrating negative outcomes. Thus, we hypothesized that identifying transcripts that are differentially regulated between waking induced by sleep deprivation and waking induced by starvation would identify genes that underlie the deleterious effects of sleep deprivation and/or protect flies from the negative consequences of waking. DESIGN We used partial complementary DNA microarrays to identify transcripts that are differentially expressed between cyc(01) mutants that had been sleep deprived or starved for 7 h. We then used genetics to determine whether disrupting genes involved in lipid metabolism would exhibit alterations in their response to sleep deprivation. SETTING Laboratory. PATIENTS OR PARTICIPANTS Drosophila melanogaster. INTERVENTIONS Sleep deprivation and starvation. MEASUREMENTS AND RESULTS We identified 84 genes with transcript levels that were differentially modulated by 7 h of sleep deprivation and starvation in cyc(01) mutants and were confirmed in independent samples using quantitative polymerase chain reaction. Several of these genes were predicted to be lipid metabolism genes, including bubblegum, cueball, and CG4500, which based on our data we have renamed heimdall (hll). Using lipidomics we confirmed that knockdown of hll using RNA interference significantly decreased lipid stores. Importantly, genetically modifying bubblegum, cueball, or hll resulted in sleep rebound alterations following sleep deprivation compared to genetic background controls. CONCLUSIONS We have identified a set of genes that may confer resilience/vulnerability to sleep deprivation and demonstrate that genes involved in lipid metabolism modulate sleep homeostasis.
Collapse
Affiliation(s)
- Matthew S Thimgan
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO.,Missouri University of Science and Technology, Department of Biological Sciences, Rolla, MO
| | - Laurent Seugnet
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO.,Centre de Recherche en Neurosciences de Lyon, Integrated Physiology of Arousal Systems Team, Lyon, France
| | - John Turk
- Division of Endocrinology, Diabetes, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Paul J Shaw
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
32
|
Genome-wide RNAi screen identifies networks involved in intestinal stem cell regulation in Drosophila. Cell Rep 2015; 10:1226-38. [PMID: 25704823 DOI: 10.1016/j.celrep.2015.01.051] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 11/06/2014] [Accepted: 01/16/2015] [Indexed: 12/17/2022] Open
Abstract
The intestinal epithelium is the most rapidly self-renewing tissue in adult animals and maintained by intestinal stem cells (ISCs) in both Drosophila and mammals. To comprehensively identify genes and pathways that regulate ISC fates, we performed a genome-wide transgenic RNAi screen in adult Drosophila intestine and identified 405 genes that regulate ISC maintenance and lineage-specific differentiation. By integrating these genes into publicly available interaction databases, we further developed functional networks that regulate ISC self-renewal, ISC proliferation, ISC maintenance of diploid status, ISC survival, ISC-to-enterocyte (EC) lineage differentiation, and ISC-to-enteroendocrine (EE) lineage differentiation. By comparing regulators among ISCs, female germline stem cells, and neural stem cells, we found that factors related to basic stem cell cellular processes are commonly required in all stem cells, and stem-cell-specific, niche-related signals are required only in the unique stem cell type. Our findings provide valuable insights into stem cell maintenance and lineage-specific differentiation.
Collapse
|
33
|
Ajjuri RR, Hall M, Reiter LT, O’Donnell JM. Drosophila. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00005-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
34
|
Giuliano MW, Miller SJ. Site-Selective Reactions with Peptide-Based Catalysts. SITE-SELECTIVE CATALYSIS 2015; 372:157-201. [DOI: 10.1007/128_2015_653] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Nakayama M, Ishibashi T, Ishikawa HO, Sato H, Usui T, Okuda T, Yashiro H, Ishikawa H, Taikou Y, Minami A, Kato K, Taki M, Aigaki T, Gunji W, Ohtsu M, Murakami Y, Tanuma SI, Tsuboi A, Adachi M, Kuroda J, Sasamura T, Yamakawa T, Matsuno K. A gain-of-function screen to identify genes that reduce lifespan in the adult of Drosophila melanogaster. BMC Genet 2014; 15:46. [PMID: 24739137 PMCID: PMC4021436 DOI: 10.1186/1471-2156-15-46] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 04/08/2014] [Indexed: 11/10/2022] Open
Abstract
Background Several lines of evidence associate misregulated genetic expression with risk factors for diabetes, Alzheimer’s, and other diseases that sporadically develop in healthy adults with no background of hereditary disorders. Thus, we are interested in genes that may be expressed normally through parts of an individual’s life, but can cause physiological defects and disease when misexpressed in adulthood. Results We attempted to identify these genes in a model organism by arbitrarily misexpressing specific genes in adult Drosophila melanogaster, using 14,133 Gene Search lines. We identified 39 “reduced-lifespan genes” that, when misexpressed in adulthood, shortened the flies’ lifespan to less than 30% of that of control flies. About half of these genes have human orthologs that are known to be involved in human diseases. For about one-fourth of the reduced-lifespan genes, suppressing apoptosis restored the lifespan shortened by their misexpression. We determined the organs responsible for reduced lifespan when these genes were misexpressed specifically in adulthood, and found that while some genes induced reduced lifespan only when misexpressed in specific adult organs, others could induce reduced lifespan when misexpressed in various organs. This finding suggests that tissue-specific dysfunction may be involved in reduced lifespan related to gene misexpression. Gene ontology analysis showed that reduced-lifespan genes are biased toward genes related to development. Conclusions We identified 39 genes that, when misexpressed in adulthood, shortened the lifespan of adult flies. Suppressing apoptosis rescued this shortened lifespan for only a subset of the reduced-lifespan genes. The adult tissues in which gene misexpression caused early death differed among the reduced-lifespan genes. These results suggest that the cause of reduced lifespan upon misexpression differed among the genes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kenji Matsuno
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
| |
Collapse
|
36
|
Abstract
Long-chain fatty acyl-coenzyme As (CoAs) are critical regulatory molecules and metabolic intermediates. The initial step in their synthesis is the activation of fatty acids by one of 13 long-chain acyl-CoA synthetase isoforms. These isoforms are regulated independently and have different tissue expression patterns and subcellular locations. Their acyl-CoA products regulate metabolic enzymes and signaling pathways, become oxidized to provide cellular energy, and are incorporated into acylated proteins and complex lipids such as triacylglycerol, phospholipids, and cholesterol esters. Their differing metabolic fates are determined by a network of proteins that channel the acyl-CoAs toward or away from specific metabolic pathways and serve as the basis for partitioning. This review evaluates the evidence for acyl-CoA partitioning by reviewing experimental data on proteins that are believed to contribute to acyl-CoA channeling, the metabolic consequences of loss of these proteins, and the potential role of maladaptive acyl-CoA partitioning in the pathogenesis of metabolic disease and carcinogenesis.
Collapse
|
37
|
Manfredini F, Riba-Grognuz O, Wurm Y, Keller L, Shoemaker D, Grozinger CM. Sociogenomics of cooperation and conflict during colony founding in the fire ant Solenopsis invicta. PLoS Genet 2013; 9:e1003633. [PMID: 23950725 PMCID: PMC3738511 DOI: 10.1371/journal.pgen.1003633] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/30/2013] [Indexed: 11/18/2022] Open
Abstract
One of the fundamental questions in biology is how cooperative and altruistic behaviors evolved. The majority of studies seeking to identify the genes regulating these behaviors have been performed in systems where behavioral and physiological differences are relatively fixed, such as in the honey bee. During colony founding in the monogyne (one queen per colony) social form of the fire ant Solenopsis invicta, newly-mated queens may start new colonies either individually (haplometrosis) or in groups (pleometrosis). However, only one queen (the “winner”) in pleometrotic associations survives and takes the lead of the young colony while the others (the “losers”) are executed. Thus, colony founding in fire ants provides an excellent system in which to examine the genes underpinning cooperative behavior and how the social environment shapes the expression of these genes. We developed a new whole genome microarray platform for S. invicta to characterize the gene expression patterns associated with colony founding behavior. First, we compared haplometrotic queens, pleometrotic winners and pleometrotic losers. Second, we manipulated pleometrotic couples in order to switch or maintain the social ranks of the two cofoundresses. Haplometrotic and pleometrotic queens differed in the expression of genes involved in stress response, aging, immunity, reproduction and lipid biosynthesis. Smaller sets of genes were differentially expressed between winners and losers. In the second experiment, switching social rank had a much greater impact on gene expression patterns than the initial/final rank. Expression differences for several candidate genes involved in key biological processes were confirmed using qRT-PCR. Our findings indicate that, in S. invicta, social environment plays a major role in the determination of the patterns of gene expression, while the queen's physiological state is secondary. These results highlight the powerful influence of social environment on regulation of the genomic state, physiology and ultimately, social behavior of animals. The characterization of the genomic basis for complex behaviors is one of the major goals of biological research. The genomic state of an individual results from the interplay between its internal condition (the “nature”) and the external environment (the “nurture”), which may include the social environment. Colony founding in the fire ant Solenopsis invicta is a complex process that serves as a useful model for investigating how the interplay between genes and social environment shapes social behavior. Unrelated, newly mated S. invicta queens may start a new colony as a group, but ultimately only one queen will survive and gain full reproductive dominance. By uncovering the genetic basis for founding behavior in fire ants we therefore provide useful insights into how cooperative behavior evolved in a context that might be considered primitively eusocial, because newly mated queens in a founding association are morphologically, physiologically and genetically very similar and display no evident division of labor. Our results suggest that social environment (founding singly or in pairs, switching dominance rank vs. maintaining rank) is a much greater driver of gene expression changes than social rank itself, suggesting that social environment, and not reproductive state, is a key regulator of gene expression, physiology and ultimately, behavior.
Collapse
Affiliation(s)
- Fabio Manfredini
- Department of Entomology, Center for Pollinator Research, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Kim M, Park HL, Park HW, Ro SH, Nam SG, Reed JM, Guan JL, Lee JH. Drosophila Fip200 is an essential regulator of autophagy that attenuates both growth and aging. Autophagy 2013; 9:1201-13. [PMID: 23819996 DOI: 10.4161/auto.24811] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Autophagy-related 1 (Atg1)/Unc-51-like protein kinases (ULKs) are evolutionarily conserved proteins that play critical physiological roles in controlling autophagy, cell growth and neurodevelopment. RB1-inducible coiled-coil 1 (RB1CC1), also known as PTK2/FAK family-interacting protein of 200 kDa (FIP200) is a recently discovered binding partner of ULK1. Here we isolated the Drosophila RB1CC1/FIP200 homolog (Fip200/CG1347) and showed that it mediates Atg1-induced autophagy as a genetically downstream component in diverse physiological contexts. Fip200 loss-of-function mutants experienced severe mobility loss associated with neuronal autophagy defects and neurodegeneration. The Fip200 mutants were also devoid of both developmental and starvation-induced autophagy in salivary gland and fat body, while having no defects in axonal transport and projection in developing neurons. Interestingly, moderate downregulation of Fip200 accelerated both developmental growth and aging, accompanied by target of rapamycin (Tor) signaling upregulation. These results suggest that Fip200 is a critical downstream component of Atg1 and specifically mediates Atg1's autophagy-, aging- and growth-regulating functions.
Collapse
Affiliation(s)
- Myungjin Kim
- Department of Molecular and Integrative Physiology; University of Michigan; Ann Arbor, MI USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Using natural variation in Drosophila to discover previously unknown endoplasmic reticulum stress genes. Proc Natl Acad Sci U S A 2013; 110:9013-8. [PMID: 23667151 DOI: 10.1073/pnas.1307125110] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Natural genetic variation is a rich resource for identifying novel elements of cellular pathways such as endoplasmic reticulum (ER) stress. ER stress occurs when misfolded proteins accumulate in the ER and cells respond with the conserved unfolded protein response (UPR), which includes large-scale gene expression changes. Although ER stress can be a cause or a modifying factor of human disease, little is known of the amount of variation in the response to ER stress and the genes contributing to such variation. To study natural variation in ER stress response in a model system, we measured the survival time in response to tunicamycin-induced ER stress in flies from 114 lines from the sequenced Drosophila Genetic Reference Panel of wild-derived inbred strains. These lines showed high heterogeneity in survival time under ER stress conditions. To identify the genes that may be driving this phenotypic variation, we profiled ER stress-induced gene expression and performed an association study. Microarray analysis identified variation in transcript levels of numerous known and previously unknown ER stress-responsive genes. Survival time was significantly associated with polymorphisms in candidate genes with known (i.e., Xbp1) and unknown roles in ER stress. Functional testing found that 17 of 25 tested candidate genes from the association study have putative roles in ER stress. In both approaches, one-third of ER stress genes had human orthologs that contribute to human disease. This study establishes Drosophila as a useful model for studying variation in ER stress and identifying ER stress genes that may contribute to human disease.
Collapse
|
40
|
Transcriptome Profiling Following Neuronal and Glial Expression of ALS-Linked SOD1 in Drosophila. G3-GENES GENOMES GENETICS 2013; 3:695-708. [PMID: 23550139 PMCID: PMC3618356 DOI: 10.1534/g3.113.005850] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) generally is a late-onset neurodegenerative disease. Mutations in the Cu/Zn superoxide dismutase 1 (SOD1) gene account for approximately 20% of familial ALS and 2% of all ALS cases. Although a number of hypotheses have been proposed to explain mutant SOD1 toxicity, the molecular mechanisms of the disease remain unclear. SOD1-linked ALS is thought to function in a non-cell-autonomous manner such that motoneurons are critical for the onset, and glia contribute to progression of the disease. Recently, it has been shown in Drosophila melanogaster that expression of human SOD1 in a subset of neuronal cells causes synaptic transmission defects, modified motor function, and altered sensitivity to compounds that induce oxidative stress. Here we used the Gal4-UAS (Upstream Activation Sequence) system to further characterize flies expressing wild-type Drosophila SOD1 (dSOD1) and the mutant human SOD1G85R (G85R) allele in motoneurons and glia. Cell-specific expression of both dSOD1 and G85R was found to influence lifespan, affect sensitivity to hydrogen peroxide, and alter lipid peroxidation levels. To better understand the genetic consequences of G85R expression in motoneurons and glia, we conducted microarray analysis of both young flies (5 days old) and old flies (45 days old) expressing G85R selectively in motoneurons or glia and concurrently in motoneurons and glia. Results from this microarray experiment identified candidate genes for further investigation and may help elucidate the individual and combined contributions of motoneurons and glia in ALS.
Collapse
|
41
|
Liu Z, Huang X. Lipid metabolism in Drosophila: development and disease. Acta Biochim Biophys Sin (Shanghai) 2013; 45:44-50. [PMID: 23257293 DOI: 10.1093/abbs/gms105] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Proteins, nucleic acids, and lipids are three major components of the cell. Despite a few basic metabolic pathways, we know very little about lipids, compared with the explosion of knowledge about proteins and nucleic acids. How many different forms of lipids are there? What are the in vivo functions of individual lipid? How does lipid metabolism contribute to normal development and human health? Many of these questions remain unanswered. For over a century, the fruit fly Drosophila melanogaster has been used as a model organism to study basic biological questions. In recent years, increasing evidences proved that Drosophila models are highly valuable for lipid metabolism and energy homeostasis researches. Some recent progresses of lipid metabolic regulation during Drosophila development and in Drosophila models of human diseases will be discussed in this review.
Collapse
Affiliation(s)
- Zhonghua Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | | |
Collapse
|
42
|
Richard FJ, Holt HL, Grozinger CM. Effects of immunostimulation on social behavior, chemical communication and genome-wide gene expression in honey bee workers (Apis mellifera). BMC Genomics 2012; 13:558. [PMID: 23072398 PMCID: PMC3483235 DOI: 10.1186/1471-2164-13-558] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 10/08/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Social insects, such as honey bees, use molecular, physiological and behavioral responses to combat pathogens and parasites. The honey bee genome contains all of the canonical insect immune response pathways, and several studies have demonstrated that pathogens can activate expression of immune effectors. Honey bees also use behavioral responses, termed social immunity, to collectively defend their hives from pathogens and parasites. These responses include hygienic behavior (where workers remove diseased brood) and allo-grooming (where workers remove ectoparasites from nestmates). We have previously demonstrated that immunostimulation causes changes in the cuticular hydrocarbon profiles of workers, which results in altered worker-worker social interactions. Thus, cuticular hydrocarbons may enable workers to identify sick nestmates, and adjust their behavior in response. Here, we test the specificity of behavioral, chemical and genomic responses to immunostimulation by challenging workers with a panel of different immune stimulants (saline, Sephadex beads and Gram-negative bacteria E. coli). RESULTS While only bacteria-injected bees elicited altered behavioral responses from healthy nestmates compared to controls, all treatments resulted in significant changes in cuticular hydrocarbon profiles. Immunostimulation caused significant changes in expression of hundreds of genes, the majority of which have not been identified as members of the canonical immune response pathways. Furthermore, several new candidate genes that may play a role in cuticular hydrocarbon biosynthesis were identified. Effects of immune challenge expression of several genes involved in immune response, cuticular hydrocarbon biosynthesis, and the Notch signaling pathway were confirmed using quantitative real-time PCR. Finally, we identified common genes regulated by pathogen challenge in honey bees and other insects. CONCLUSIONS These results demonstrate that honey bee genomic responses to immunostimulation are substantially broader than the previously identified canonical immune response pathways, and may mediate the behavioral changes associated with social immunity by orchestrating changes in chemical signaling. These studies lay the groundwork for future research into the genomic responses of honey bees to native honey bee parasites and pathogens.
Collapse
Affiliation(s)
- Freddie-Jeanne Richard
- Laboratoire Ecologie Evolution Symbiose, UMR CNRS 6556, University of Poitiers, Cedex, POITIERS, France.
| | | | | |
Collapse
|
43
|
Newman T, Sinadinos C, Johnston A, Sealey M, Mudher A. Using Drosophila models of neurodegenerative diseases for drug discovery. Expert Opin Drug Discov 2012; 6:129-40. [PMID: 22647132 DOI: 10.1517/17460441.2011.549124] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and Huntington's disease are increasing in prevalence as our aging population increases in size. Despite this, currently there are no disease-modifying drugs available for the treatment of these conditions. Drosophila melanogaster is a highly tractable model organism that has been successfully used to emulate various aspects of these diseases in vivo. These Drosophila models have not been fully exploited in drug discovery and design strategies. AREAS COVERED This review explores how Drosophila models can be used to facilitate drug discovery. Specifically, we review their uses as a physiologically-relevant medium to high-throughput screening tool for the identification of therapeutic compounds and discuss how they can aid drug discovery by highlighting disease mechanisms that may serve as druggable targets in the future. The reader will appreciate how the various attributes of Drosophila make it an unsurpassed model organism and how Drosophila models of neurodegeneration can contribute to drug discovery in a variety of ways. EXPERT OPINION Drosophila models of human neurodegenerative diseases can make a significant contribution to the unmet need of disease-modifying therapeutic intervention for the treatment of these increasingly common neurodegenerative conditions.
Collapse
Affiliation(s)
- Tracey Newman
- University of Southampton, School of Medicine, Life Science Building 85, Southampton, SO17 1BJ, UK +44 2380 597642 ;
| | | | | | | | | |
Collapse
|
44
|
Faust JE, Verma A, Peng C, McNew JA. An inventory of peroxisomal proteins and pathways in Drosophila melanogaster. Traffic 2012; 13:1378-92. [PMID: 22758915 PMCID: PMC3443258 DOI: 10.1111/j.1600-0854.2012.01393.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 06/28/2012] [Accepted: 07/03/2012] [Indexed: 11/29/2022]
Abstract
Peroxisomes are ubiquitous organelles housing a variety of essential biochemical pathways. Peroxisome dysfunction causes a spectrum of human diseases known as peroxisome biogenesis disorders (PBD). Although much is known regarding the mechanism of peroxisome biogenesis, it is still unclear how peroxisome dysfunction leads to the disease state. Several recent studies have shown that mutations in Drosophila peroxin genes cause phenotypes similar to those seen in humans with PBDs suggesting that Drosophila might be a useful system to model PBDs. We have analyzed the proteome of Drosophila to identify the proteins involved in peroxisomal biogenesis and homeostasis as well as metabolic enzymes that function within the organelle. The subcellular localization of five of these predicted peroxisomal proteins was confirmed. Similar to Caenorhabditis elegans, Drosophila appears to only utilize the peroxisome targeting signal type 1 system for matrix protein import. This work will further our understanding of peroxisomes in Drosophila and add to the usefulness of this emerging model system.
Collapse
Affiliation(s)
- Joseph E. Faust
- Department of Biochemistry and Cell Biology, Rice University, 6100 Main Street MS601, Houston, TX 77005
| | - Avani Verma
- Department of Biochemistry and Cell Biology, Rice University, 6100 Main Street MS601, Houston, TX 77005
| | - Chengwei Peng
- Department of Biochemistry and Cell Biology, Rice University, 6100 Main Street MS601, Houston, TX 77005
| | - James A. McNew
- Department of Biochemistry and Cell Biology, Rice University, 6100 Main Street MS601, Houston, TX 77005
| |
Collapse
|
45
|
Miranda CO, Brites P, Mendes Sousa M, Teixeira CA. Advances and pitfalls of cell therapy in metabolic leukodystrophies. Cell Transplant 2012; 22:189-204. [PMID: 23006656 DOI: 10.3727/096368912x656117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Leukodystrophies are a group of disorders characterized by myelin dysfunction, either at the level of myelin formation or maintenance, that affect the central nervous system (CNS) and also in some cases, to a lesser extent, the peripheral nervous system (PNS). Although these genetic-based disorders are generally rare, all together they have a significant impact in the society, with an estimated overall incidence of 1 in 7,663 live births. Currently, there is no cure for leukodystrophies, and the development of effective treatments remains challenging. Not only leukodystrophies generally progress very fast, but also most are multifocal needing the simultaneous targeting at multiple sites. Moreover, as the CNS is affected, the blood-brain barrier (BBB) limits the efficacy of treatment. Recently, interest on cell therapy has increased, and the leukodystrophies for which metabolic correction is needed have become first-choice candidates for cell-based clinical trials. In this review, we present and discuss the available cell transplantation therapies in metabolic leukodystrophies including fucosidosis, X-linked adrenoleukodystrophy, metachromatic leukodystrophy, Canavan disease, and Krabbe's disease. We will discuss the latest advances of cell therapy and its pitfalls in this group of disorders, taking into account, among others, the limitations imposed by reduced cell migration in multifocal conditions, the need to achieve corrective enzyme threshold levels, and the growing awareness that not only myelin but also the associated axonopathy needs to be targeted in some leukodystrophies.
Collapse
|
46
|
Jaiswal M, Sandoval H, Zhang K, Bayat V, Bellen HJ. Probing mechanisms that underlie human neurodegenerative diseases in Drosophila. Annu Rev Genet 2012; 46:371-96. [PMID: 22974305 DOI: 10.1146/annurev-genet-110711-155456] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The fruit fly, Drosophila melanogaster, is an excellent organism for the study of the genetic and molecular basis of metazoan development. Drosophila provides numerous tools and reagents to unravel the molecular and cellular functions of genes that cause human disease, and the past decade has witnessed a significant expansion of the study of neurodegenerative disease mechanisms in flies. Here we review the interplay between oxidative stress and neuronal toxicity. We cover some of the studies that show how proteasome degradation of protein aggregates, autophagy, mitophagy, and lysosomal function affect the quality control mechanisms required for neuronal survival. We discuss how forward genetic screens in flies have led to the isolation of a few loci that cause neurodegeneration, paving the way for large-scale systematic screens to identify such loci in flies as well as promoting gene discovery in humans.
Collapse
Affiliation(s)
- M Jaiswal
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
47
|
Uesugi M. [Control and analysis of cells by synthetic small molecules]. YAKUGAKU ZASSHI 2012; 132:575-86. [PMID: 22687693 DOI: 10.1248/yakushi.132.575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In human history, small organic molecules have been utilized for improving human health and for revealing secrets of life. Discovery or design of small organic molecules with unique biological activity permits small-molecule-initiated exploration of biology. Our laboratory has been discovering and designing bioactive small synthetic molecules to use them as tools to analyze or modulate biological processes. This personal perspective summarizes our contributions to chemical biology, particularly in the fields of gene transcription, cell therapy, growth factor signaling, and target identification.
Collapse
Affiliation(s)
- Motonari Uesugi
- Institutes for Integrated Cell-Material Sciences and for Chemical Research, Kyoto University, Japan.
| |
Collapse
|
48
|
Rudrapatna VA, Cagan RL, Das TK. Drosophila cancer models. Dev Dyn 2011; 241:107-18. [PMID: 22038952 DOI: 10.1002/dvdy.22771] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2011] [Indexed: 01/20/2023] Open
Abstract
Cancer is driven by complex genetic and cellular mechanisms. Recently, the Drosophila community has become increasingly interested in exploring cancer issues. The Drosophila field has made seminal contributions to many of the mechanisms that are fundamental to the cancer process; several of these mechanisms have already been validated in vertebrates. Less well known are the Drosophila field's early direct contributions to the cancer field: some of the earliest tumor suppressors were identified in flies. In this review, we identify major contributions that Drosophila studies have made toward dissecting the pathways and mechanisms underlying tumor progression. We also highlight areas, such as drug discovery, where we expect Drosophila studies to make a major scientific impact in the future.
Collapse
Affiliation(s)
- Vivek A Rudrapatna
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | |
Collapse
|
49
|
Schulte J, Sepp KJ, Wu C, Hong P, Littleton JT. High-content chemical and RNAi screens for suppressors of neurotoxicity in a Huntington's disease model. PLoS One 2011; 6:e23841. [PMID: 21909362 PMCID: PMC3166080 DOI: 10.1371/journal.pone.0023841] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 07/26/2011] [Indexed: 11/27/2022] Open
Abstract
To identify Huntington's Disease therapeutics, we conducted high-content small molecule and RNAi suppressor screens using a Drosophila primary neural culture Huntingtin model. Drosophila primary neurons offer a sensitive readout for neurotoxicty, as their neurites develop dysmorphic features in the presence of mutant polyglutamine-expanded Huntingtin compared to nonpathogenic Huntingtin. By tracking the subcellular distribution of mRFP-tagged pathogenic Huntingtin and assaying neurite branch morphology via live-imaging, we identified suppressors that could reduce Huntingtin aggregation and/or prevent the formation of dystrophic neurites. The custom algorithms we used to quantify neurite morphologies in complex cultures provide a useful tool for future high-content screening approaches focused on neurodegenerative disease models. Compounds previously found to be effective aggregation inhibitors in mammalian systems were also effective in Drosophila primary cultures, suggesting translational capacity between these models. However, we did not observe a direct correlation between the ability of a compound or gene knockdown to suppress aggregate formation and its ability to rescue dysmorphic neurites. Only a subset of aggregation inhibitors could revert dysmorphic cellular profiles. We identified lkb1, an upstream kinase in the mTOR/Insulin pathway, and four novel drugs, Camptothecin, OH-Camptothecin, 18β-Glycyrrhetinic acid, and Carbenoxolone, that were strong suppressors of mutant Huntingtin-induced neurotoxicity. Huntingtin neurotoxicity suppressors identified through our screen also restored viability in an in vivo Drosophila Huntington's Disease model, making them attractive candidates for further therapeutic evaluation.
Collapse
Affiliation(s)
- Joost Schulte
- Department of Biology, Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America.
| | | | | | | | | |
Collapse
|
50
|
Mating alters gene expression patterns in Drosophila melanogaster male heads. BMC Genomics 2010; 11:558. [PMID: 20937114 PMCID: PMC3091707 DOI: 10.1186/1471-2164-11-558] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 10/11/2010] [Indexed: 11/28/2022] Open
Abstract
Background Behavior is a complex process resulting from the integration of genetic and environmental information. Drosophila melanogaster rely on multiple sensory modalities for reproductive success, and mating causes physiological changes in both sexes that affect reproductive output or behavior. Some of these effects are likely mediated by changes in gene expression. Courtship and mating alter female transcript profiles, but it is not known how mating affects male gene expression. Results We used Drosophila genome arrays to identify changes in gene expression profiles that occur in mated male heads. Forty-seven genes differed between mated and control heads 2 hrs post mating. Many mating-responsive genes are highly expressed in non-neural head tissues, including an adipose tissue called the fat body. One fat body-enriched gene, female-specific independent of transformer (fit), is a downstream target of the somatic sex-determination hierarchy, a genetic pathway that regulates Drosophila reproductive behaviors as well as expression of some fat-expressed genes; three other mating-responsive loci are also downstream components of this pathway. Another mating-responsive gene expressed in fat, Juvenile hormone esterase (Jhe), is necessary for robust male courtship behavior and mating success. Conclusions Our study demonstrates that mating causes changes in male head gene expression profiles and supports an increasing body of work implicating adipose signaling in behavior modulation. Since several mating-induced genes are sex-determination hierarchy target genes, additional mating-responsive loci may be downstream components of this pathway as well.
Collapse
|