1
|
Vandermeulen MD, Khaiwal S, Rubio G, Liti G, Cullen PJ. Gain- and loss-of-function alleles within signaling pathways lead to phenotypic diversity among individuals. iScience 2024; 27:110860. [PMID: 39381740 PMCID: PMC11460476 DOI: 10.1016/j.isci.2024.110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/29/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024] Open
Abstract
Understanding how phenotypic diversity is generated is an important question in biology. We explored phenotypic diversity among wild yeast isolates (Saccharomyces cerevisiae) and found variation in the activity of MAPK signaling pathways as a contributing mechanism. To uncover the genetic basis of this mechanism, we identified 1957 SNPs in 62 candidate genes encoding signaling proteins from a MAPK signaling module within a large collection of yeast (>1500 individuals). Follow-up testing identified functionally relevant variants in key signaling proteins. Loss-of-function (LOF) alleles in a PAK kinase impacted protein stability and pathway specificity decreasing filamentous growth and mating phenotypes. In contrast, gain-of-function (GOF) alleles in G-proteins that were hyperactivating induced filamentous growth. Similar amino acid substitutions in G-proteins were identified in metazoans that in some cases were fixed in multicellular lineages including humans, suggesting hyperactivating GOF alleles may play roles in generating phenotypic diversity across eukaryotes. A mucin signaler that regulates MAPK activity was also found to contain a prevalance of presumed GOF alleles amoung individuals based on changes in mucin repeat numbers. Thus, genetic variation in signaling pathways may act as a reservoir for generating phenotypic diversity across eukaryotes.
Collapse
Affiliation(s)
| | - Sakshi Khaiwal
- Université Côte d’Azur, CNRS, INSERM, IRCAN, Nice, France
| | - Gabriel Rubio
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-1300, USA
| | - Gianni Liti
- Université Côte d’Azur, CNRS, INSERM, IRCAN, Nice, France
| | - Paul J. Cullen
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-1300, USA
| |
Collapse
|
2
|
Vandermeulen MD, Lorenz MC, Cullen PJ. Conserved signaling modules regulate filamentous growth in fungi: a model for eukaryotic cell differentiation. Genetics 2024; 228:iyae122. [PMID: 39239926 PMCID: PMC11457945 DOI: 10.1093/genetics/iyae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/20/2024] [Indexed: 09/07/2024] Open
Abstract
Eukaryotic organisms are composed of different cell types with defined shapes and functions. Specific cell types are produced by the process of cell differentiation, which is regulated by signal transduction pathways. Signaling pathways regulate cell differentiation by sensing cues and controlling the expression of target genes whose products generate cell types with specific attributes. In studying how cells differentiate, fungi have proved valuable models because of their ease of genetic manipulation and striking cell morphologies. Many fungal species undergo filamentous growth-a specialized growth pattern where cells produce elongated tube-like projections. Filamentous growth promotes expansion into new environments, including invasion into plant and animal hosts by fungal pathogens. The same signaling pathways that regulate filamentous growth in fungi also control cell differentiation throughout eukaryotes and include highly conserved mitogen-activated protein kinase (MAPK) pathways, which is the focus of this review. In many fungal species, mucin-type sensors regulate MAPK pathways to control filamentous growth in response to diverse stimuli. Once activated, MAPK pathways reorganize cell polarity, induce changes in cell adhesion, and promote the secretion of degradative enzymes that mediate access to new environments. However, MAPK pathway regulation is complicated because related pathways can share components with each other yet induce unique responses (i.e. signal specificity). In addition, MAPK pathways function in highly integrated networks with other regulatory pathways (i.e. signal integration). Here, we discuss signal specificity and integration in several yeast models (mainly Saccharomyces cerevisiae and Candida albicans) by focusing on the filamentation MAPK pathway. Because of the strong evolutionary ties between species, a deeper understanding of the regulation of filamentous growth in established models and increasingly diverse fungal species can reveal fundamentally new mechanisms underlying eukaryotic cell differentiation.
Collapse
Affiliation(s)
| | - Michael C Lorenz
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Paul J Cullen
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-1300, USA
| |
Collapse
|
3
|
Luo Z, Xiong D, Tian C. The Roles of Gti1/Pac2 Family Proteins in Fungal Growth, Morphogenesis, Stress Response, and Pathogenicity. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2024; 37:488-497. [PMID: 38427716 DOI: 10.1094/mpmi-11-23-0198-cr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Gti1/Pac2 is a fungal-specific transcription factor family with a stable and conserved N-terminal domain. Generally, there are two members in this family, named Gti1/Wor1/Rpy1/Mit1/Reg1/Ros1/Sge1 and Pac2, which are involved in fungal growth, development, stress response, spore production, pathogenicity, and so on. The Gti1/Pac2 family proteins share some conserved and distinct functions. For example, in Schizosaccharomyces pombe, Gti1 promotes the initiation of gluconate uptake during glucose starvation, while Pac2 controls the onset of sexual development in a pathway independent of the cAMP cascade. In the last two decades, more attention was focused on the Gti1 and its orthologs because of their significant effect on morphological switching and fungal virulence. By contrast, limited work was published on the functions of Pac2, which is required for stress responses and conidiation, but plays a minor role in fungal virulence. In this review, we present an overview of our current understanding of the Gti1/Pac2 proteins that contribute to fungal development and/or pathogenicity and of the regulation mechanisms during infection related development. Understanding the working networks of the conserved Gti1/Pac2 transcription factors in fungal pathogenicity not only advances our knowledge of the highly elaborate infection process but may also lead to the development of novel strategies for the control of plant disease. [Formula: see text] Copyright © 2024 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.
Collapse
Affiliation(s)
- Zheng Luo
- State Key laboratory of Efficient Production of Forest Resources, Beijing Forestry University, Beijing 100083, China
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
- The Key Laboratory for Silviculture and Conservation of Ministry of Education, College of Forestry, Beijing Forestry University, Beijing 100083, China
| | - Dianguang Xiong
- State Key laboratory of Efficient Production of Forest Resources, Beijing Forestry University, Beijing 100083, China
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
- The Key Laboratory for Silviculture and Conservation of Ministry of Education, College of Forestry, Beijing Forestry University, Beijing 100083, China
| | - Chengming Tian
- State Key laboratory of Efficient Production of Forest Resources, Beijing Forestry University, Beijing 100083, China
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
- The Key Laboratory for Silviculture and Conservation of Ministry of Education, College of Forestry, Beijing Forestry University, Beijing 100083, China
| |
Collapse
|
4
|
Vandermeulen MD, Cullen PJ. Ecological inducers of the yeast filamentous growth pathway reveal environment-dependent roles for pathway components. mSphere 2023; 8:e0028423. [PMID: 37732804 PMCID: PMC10597418 DOI: 10.1128/msphere.00284-23] [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: 05/25/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Signaling modules, such as mitogen-activated protein kinase (MAPK) pathways, are evolutionarily conserved drivers of cell differentiation and stress responses. In many fungal species including pathogens, MAPK pathways control filamentous growth, where cells differentiate into an elongated cell type. The convenient model budding yeast Saccharomyces cerevisiae undergoes filamentous growth by the filamentous growth (fMAPK) pathway; however, the inducers of the pathway remain unclear, perhaps because pathway activity has been mainly studied in laboratory conditions. To address this knowledge gap, an ecological framework was used, which uncovered new fMAPK pathway inducers, including pectin, a material found in plants, and the metabolic byproduct ethanol. We also show that induction by a known inducer of the pathway, the non-preferred carbon source galactose, required galactose metabolism and induced the pathway differently than glucose limitation or other non-preferred carbon sources. By exploring fMAPK pathway function in fruit, we found that induction of the pathway led to visible digestion of fruit rind through a known target, PGU1, which encodes a pectolytic enzyme. Combinations of inducers (galactose and ethanol) stimulated the pathway to near-maximal levels, which showed dispensability of several fMAPK pathway components (e.g., mucin sensor, p21-activated kinase), but not others (e.g., adaptor, MAPKKK) and required the Ras2-protein kinase A pathway. This included a difference between the transcription factor binding partners for the pathway, as Tec1p, but not Ste12p, was partly dispensable for fMAPK pathway activity. Thus, by exploring ecologically relevant stimuli, new modes of MAPK pathway signaling were uncovered, perhaps revealing how a pathway can respond differently to specific environments. IMPORTANCE Filamentous growth is a cell differentiation response and important aspect of fungal biology. In plant and animal fungal pathogens, filamentous growth contributes to virulence. One signaling pathway that regulates filamentous growth is an evolutionarily conserved MAPK pathway. The yeast Saccharomyces cerevisiae is a convenient model to study MAPK-dependent regulation of filamentous growth, although the inducers of the pathway are not clear. Here, we exposed yeast cells to ecologically relevant compounds (e.g., plant compounds), which identified new inducers of the MAPK pathway. In combination, the inducers activated the pathway to near-maximal levels but did not cause detrimental phenotypes associated with previously identified hyperactive alleles. This context allowed us to identify conditional bypass for multiple pathway components. Thus, near-maximal induction of a MAPK pathway by ecologically relevant inducers provides a powerful tool to assess cellular signaling during a fungal differentiation response.
Collapse
Affiliation(s)
| | - Paul J. Cullen
- Department of Biological Sciences, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
5
|
Vandermeulen MD, Cullen PJ. Gene by Environment Interactions reveal new regulatory aspects of signaling network plasticity. PLoS Genet 2022; 18:e1009988. [PMID: 34982769 PMCID: PMC8759647 DOI: 10.1371/journal.pgen.1009988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 01/14/2022] [Accepted: 12/09/2021] [Indexed: 11/18/2022] Open
Abstract
Phenotypes can change during exposure to different environments through the regulation of signaling pathways that operate in integrated networks. How signaling networks produce different phenotypes in different settings is not fully understood. Here, Gene by Environment Interactions (GEIs) were used to explore the regulatory network that controls filamentous/invasive growth in the yeast Saccharomyces cerevisiae. GEI analysis revealed that the regulation of invasive growth is decentralized and varies extensively across environments. Different regulatory pathways were critical or dispensable depending on the environment, microenvironment, or time point tested, and the pathway that made the strongest contribution changed depending on the environment. Some regulators even showed conditional role reversals. Ranking pathways' roles across environments revealed an under-appreciated pathway (OPI1) as the single strongest regulator among the major pathways tested (RAS, RIM101, and MAPK). One mechanism that may explain the high degree of regulatory plasticity observed was conditional pathway interactions, such as conditional redundancy and conditional cross-pathway regulation. Another mechanism was that different pathways conditionally and differentially regulated gene expression, such as target genes that control separate cell adhesion mechanisms (FLO11 and SFG1). An exception to decentralized regulation of invasive growth was that morphogenetic changes (cell elongation and budding pattern) were primarily regulated by one pathway (MAPK). GEI analysis also uncovered a round-cell invasion phenotype. Our work suggests that GEI analysis is a simple and powerful approach to define the regulatory basis of complex phenotypes and may be applicable to many systems.
Collapse
Affiliation(s)
- Matthew D. Vandermeulen
- Department of Biological Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Paul J. Cullen
- Department of Biological Sciences, University at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
6
|
Qin J, Hu Y, Yao JC, Leung RWT, Zhou Y, Qin Y, Wang J. Cell fate conversion prediction by group sparse optimization method utilizing single-cell and bulk OMICs data. Brief Bioinform 2021; 22:6347206. [PMID: 34374760 DOI: 10.1093/bib/bbab311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/06/2021] [Accepted: 07/19/2021] [Indexed: 01/09/2023] Open
Abstract
Cell fate conversion by overexpressing defined factors is a powerful tool in regenerative medicine. However, identifying key factors for cell fate conversion requires laborious experimental efforts; thus, many of such conversions have not been achieved yet. Nevertheless, cell fate conversions found in many published studies were incomplete as the expression of important gene sets could not be manipulated thoroughly. Therefore, the identification of master transcription factors for complete and efficient conversion is crucial to render this technology more applicable clinically. In the past decade, systematic analyses on various single-cell and bulk OMICs data have uncovered numerous gene regulatory mechanisms, and made it possible to predict master gene regulators during cell fate conversion. By virtue of the sparse structure of master transcription factors and the group structure of their simultaneous regulatory effects on the cell fate conversion process, this study introduces a novel computational method predicting master transcription factors based on group sparse optimization technique integrating data from multi-OMICs levels, which can be applicable to both single-cell and bulk OMICs data with a high tolerance of data sparsity. When it is compared with current prediction methods by cross-referencing published and validated master transcription factors, it possesses superior performance. In short, this method facilitates fast identification of key regulators, give raise to the possibility of higher successful conversion rate and in the hope of reducing experimental cost.
Collapse
Affiliation(s)
- Jing Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaohua Hu
- Shenzhen Key Laboratory of Advanced Machine Learning and Applications, College of Mathematics and Statistics, Shenzhen University, Shenzhen 518060, China
| | - Jen-Chih Yao
- Research Center for Interneural Computing, China Medical University, Taichung 40402, Taiwan
| | - Ricky Wai Tak Leung
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yongqiang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yiming Qin
- Center for Genomic Sciences & School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Junwen Wang
- Department of Quantitative Health Sciences and Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA.,Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ 85259, USA
| |
Collapse
|
7
|
Buchberger E, Bilen A, Ayaz S, Salamanca D, Matas de las Heras C, Niksic A, Almudi I, Torres-Oliva M, Casares F, Posnien N. Variation in Pleiotropic Hub Gene Expression Is Associated with Interspecific Differences in Head Shape and Eye Size in Drosophila. Mol Biol Evol 2021; 38:1924-1942. [PMID: 33386848 PMCID: PMC8097299 DOI: 10.1093/molbev/msaa335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Revealing the mechanisms underlying the breathtaking morphological diversity observed in nature is a major challenge in Biology. It has been established that recurrent mutations in hotspot genes cause the repeated evolution of morphological traits, such as body pigmentation or the gain and loss of structures. To date, however, it remains elusive whether hotspot genes contribute to natural variation in the size and shape of organs. As natural variation in head morphology is pervasive in Drosophila, we studied the molecular and developmental basis of differences in compound eye size and head shape in two closely related Drosophila species. We show differences in the progression of retinal differentiation between species and we applied comparative transcriptomics and chromatin accessibility data to identify the GATA transcription factor Pannier (Pnr) as central factor associated with these differences. Although the genetic manipulation of Pnr affected multiple aspects of dorsal head development, the effect of natural variation is restricted to a subset of the phenotypic space. We present data suggesting that this developmental constraint is caused by the coevolution of expression of pnr and its cofactor u-shaped (ush). We propose that natural variation in expression or function of highly connected developmental regulators with pleiotropic functions is a major driver for morphological evolution and we discuss implications on gene regulatory network evolution. In comparison to previous findings, our data strongly suggest that evolutionary hotspots are not the only contributors to the repeated evolution of eye size and head shape in Drosophila.
Collapse
Affiliation(s)
- Elisa Buchberger
- Department of Developmental Biology, University of Göttingen, Göttingen, Germany
| | - Anıl Bilen
- Department of Developmental Biology, University of Göttingen, Göttingen, Germany
| | - Sanem Ayaz
- Department of Developmental Biology, University of Göttingen, Göttingen, Germany
| | - David Salamanca
- Department of Developmental Biology, University of Göttingen, Göttingen, Germany
- Present address: Department of Integrative Zoology, University of Vienna, Vienna, Austria
| | | | - Armin Niksic
- Department of Developmental Biology, University of Göttingen, Göttingen, Germany
| | - Isabel Almudi
- CABD (CSIC/UPO/JA), DMC2 Unit, Pablo de Olavide University Campus, Seville, Spain
| | - Montserrat Torres-Oliva
- Department of Developmental Biology, University of Göttingen, Göttingen, Germany
- Present address: Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Fernando Casares
- CABD (CSIC/UPO/JA), DMC2 Unit, Pablo de Olavide University Campus, Seville, Spain
| | - Nico Posnien
- Department of Developmental Biology, University of Göttingen, Göttingen, Germany
- Corresponding author: E-mail:
| |
Collapse
|
8
|
Kumar A. The Complex Genetic Basis and Multilayered Regulatory Control of Yeast Pseudohyphal Growth. Annu Rev Genet 2021; 55:1-21. [PMID: 34280314 DOI: 10.1146/annurev-genet-071719-020249] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Eukaryotic cells are exquisitely responsive to external and internal cues, achieving precise control of seemingly diverse growth processes through a complex interplay of regulatory mechanisms. The budding yeast Saccharomyces cerevisiae provides a fascinating model of cell growth in its stress-responsive transition from planktonic single cells to a filamentous pseudohyphal growth form. During pseudohyphal growth, yeast cells undergo changes in morphology, polarity, and adhesion to form extended and invasive multicellular filaments. This pseudohyphal transition has been studied extensively as a model of conserved signaling pathways regulating cell growth and for its relevance in understanding the pathogenicity of the related opportunistic fungus Candida albicans, wherein filamentous growth is required for virulence. This review highlights the broad gene set enabling yeast pseudohyphal growth, signaling pathways that regulate this process, the role and regulation of proteins conferring cell adhesion, and interesting regulatory mechanisms enabling the pseudohyphal transition. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Anuj Kumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA;
| |
Collapse
|
9
|
Weidner FM, Schwab JD, Werle SD, Ikonomi N, Lausser L, Kestler HA. Capturing dynamic relevance in Boolean networks using graph theoretical measures. Bioinformatics 2021; 37:3530-3537. [PMID: 33983406 PMCID: PMC8545349 DOI: 10.1093/bioinformatics/btab277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 11/14/2022] Open
Abstract
Motivation Interaction graphs are able to describe regulatory dependencies between compounds without capturing dynamics. In contrast, mathematical models that are based on interaction graphs allow to investigate the dynamics of biological systems. However, since dynamic complexity of these models grows exponentially with their size, exhaustive analyses of the dynamics and consequently screening all possible interventions eventually becomes infeasible. Thus, we designed an approach to identify dynamically relevant compounds based on the static network topology. Results Here, we present a method only based on static properties to identify dynamically influencing nodes. Coupling vertex betweenness and determinative power, we could capture relevant nodes for changing dynamics with an accuracy of 75% in a set of 35 published logical models. Further analyses of the selected compounds’ connectivity unravelled a new class of not highly connected nodes with high impact on the networks’ dynamics, which we call gatekeepers. We validated our method’s working concept on logical models, which can be readily scaled up to complex interaction networks, where dynamic analyses are not even feasible. Availability and implementation Code is freely available at https://github.com/sysbio-bioinf/BNStatic. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Felix M Weidner
- Institute of Medical Systems Biology, Ulm University, Germany.,International Graduate School of Molecular Medicine, Ulm University, Germany
| | - Julian D Schwab
- Institute of Medical Systems Biology, Ulm University, Germany
| | - Silke D Werle
- Institute of Medical Systems Biology, Ulm University, Germany.,International Graduate School of Molecular Medicine, Ulm University, Germany
| | - Nensi Ikonomi
- Institute of Medical Systems Biology, Ulm University, Germany.,International Graduate School of Molecular Medicine, Ulm University, Germany
| | - Ludwig Lausser
- Institute of Medical Systems Biology, Ulm University, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Germany
| |
Collapse
|
10
|
Gedman G, Haase B, Durieux G, Biegler MT, Fedrigo O, Jarvis ED. As above, so below: Whole transcriptome profiling demonstrates strong molecular similarities between avian dorsal and ventral pallial subdivisions. J Comp Neurol 2021; 529:3222-3246. [PMID: 33871048 PMCID: PMC8251894 DOI: 10.1002/cne.25159] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022]
Abstract
Over the last two decades, beginning with the Avian Brain Nomenclature Forum in 2000, major revisions have been made to our understanding of the organization and nomenclature of the avian brain. However, there are still unresolved questions on avian pallial organization, particularly whether the cells above the vestigial ventricle represent distinct populations to those below it or similar populations. To test these two hypotheses, we profiled the transcriptomes of the major avian pallial subdivisions dorsal and ventral to the vestigial ventricle boundary using RNA sequencing and a new zebra finch genome assembly containing about 22,000 annotated, complete genes. We found that the transcriptomes of neural populations above and below the ventricle were remarkably similar. Each subdivision in dorsal pallium (Wulst) had a corresponding molecular counterpart in the ventral pallium (dorsal ventricular ridge). In turn, each corresponding subdivision exhibited shared gene co‐expression modules that contained gene sets enriched in functional specializations, such as anatomical structure development, synaptic transmission, signaling, and neurogenesis. These findings are more in line with the continuum hypothesis of avian brain subdivision organization above and below the vestigial ventricle space, with the pallium as a whole consisting of four major cell populations (intercalated pallium, mesopallium, hyper‐nidopallium, and arcopallium) instead of seven (hyperpallium apicale, interstitial hyperpallium apicale, intercalated hyperpallium, hyperpallium densocellare, mesopallium, nidopallium, and arcopallium). We suggest adopting a more streamlined hierarchical naming system that reflects the robust similarities in gene expression, neural connectivity motifs, and function. These findings have important implications for our understanding of overall vertebrate brain evolution.
Collapse
Affiliation(s)
- Gregory Gedman
- Laboratory of the Neurogenetics of Language, The Rockefeller University, New York, New York, USA
| | - Bettina Haase
- Laboratory of the Neurogenetics of Language, The Rockefeller University, New York, New York, USA.,Vertebrate Genome Laboratory, The Rockefeller University, New York, New York, USA
| | - Gillian Durieux
- Behavioural Genomics, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Matthew T Biegler
- Laboratory of the Neurogenetics of Language, The Rockefeller University, New York, New York, USA
| | - Olivier Fedrigo
- Laboratory of the Neurogenetics of Language, The Rockefeller University, New York, New York, USA.,Vertebrate Genome Laboratory, The Rockefeller University, New York, New York, USA
| | - Erich D Jarvis
- Laboratory of the Neurogenetics of Language, The Rockefeller University, New York, New York, USA.,Vertebrate Genome Laboratory, The Rockefeller University, New York, New York, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
11
|
Sunyer-Figueres M, Vázquez J, Mas A, Torija MJ, Beltran G. Transcriptomic Insights into the Effect of Melatonin in Saccharomyces cerevisiae in the Presence and Absence of Oxidative Stress. Antioxidants (Basel) 2020; 9:E947. [PMID: 33019712 PMCID: PMC7650831 DOI: 10.3390/antiox9100947] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022] Open
Abstract
Melatonin is a ubiquitous indolamine that plays important roles in various aspects of biological processes in mammals. In Saccharomyces cerevisiae, melatonin has been reported to exhibit antioxidant properties and to modulate the expression of some genes involved in endogenous defense systems. The aim of this study was to elucidate the role of supplemented melatonin at the transcriptional level in S. cerevisiae in the presence and absence of oxidative stress. This was achieved by exposing yeast cells pretreated with different melatonin concentrations to hydrogen peroxide and assessing the entry of melatonin into the cell and the yeast response at the transcriptional level (by microarray and qPCR analyses) and the physiological level (by analyzing changes in the lipid composition and mitochondrial activity). We found that exogenous melatonin crossed cellular membranes at nanomolar concentrations and modulated the expression of many genes, mainly downregulating the expression of mitochondrial genes in the absence of oxidative stress, triggering a hypoxia-like response, and upregulating them under stress, mainly the cytochrome complex and electron transport chain. Other categories that were enriched by the effect of melatonin were related to transport, antioxidant activity, signaling, and carbohydrate and lipid metabolism. The overall results suggest that melatonin is able to reprogram the cellular machinery to achieve tolerance to oxidative stress.
Collapse
Affiliation(s)
| | | | | | - María-Jesús Torija
- Departament de Bioquímica i Biotecnologia, Grup de Biotecnologia Enològica, Facultat d’Enologia, Universitat Rovira i Virgili, C/Marcel·lí Domingo, 1. 43007 Tarragona, Catalunya, Spain; (M.S.-F.); (J.V.); (A.M.); (G.B.)
| | | |
Collapse
|
12
|
Vandermeulen MD, Cullen PJ. New Aspects of Invasive Growth Regulation Identified by Functional Profiling of MAPK Pathway Targets in Saccharomyces cerevisiae. Genetics 2020; 216:95-116. [PMID: 32665277 PMCID: PMC7463291 DOI: 10.1534/genetics.120.303369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
MAPK pathways are drivers of morphogenesis and stress responses in eukaryotes. A major function of MAPK pathways is the transcriptional induction of target genes, which produce proteins that collectively generate a cellular response. One approach to comprehensively understand how MAPK pathways regulate cellular responses is to characterize the individual functions of their transcriptional targets. Here, by examining uncharacterized targets of the MAPK pathway that positively regulates filamentous growth in Saccharomyces cerevisiae (fMAPK pathway), we identified a new role for the pathway in negatively regulating invasive growth. Specifically, four targets were identified that had an inhibitory role in invasive growth: RPI1, RGD2, TIP1, and NFG1/YLR042cNFG1 was a highly induced unknown open reading frame that negatively regulated the filamentous growth MAPK pathway. We also identified SFG1, which encodes a transcription factor, as a target of the fMAPK pathway. Sfg1p promoted cell adhesion independently from the fMAPK pathway target and major cell adhesion flocculin Flo11p, by repressing genes encoding presumptive cell-wall-degrading enzymes. Sfg1p also contributed to FLO11 expression. Sfg1p and Flo11p regulated different aspects of cell adhesion, and their roles varied based on the environment. Sfg1p also induced an elongated cell morphology, presumably through a cell-cycle delay. Thus, the fMAPK pathway coordinates positive and negative regulatory proteins to fine-tune filamentous growth resulting in a nuanced response. Functional analysis of other pathways' targets may lead to a more comprehensive understanding of how signaling cascades generate biological responses.
Collapse
Affiliation(s)
| | - Paul J Cullen
- Department of Biological Sciences, University at Buffalo, New York 14260-1300
| |
Collapse
|
13
|
XPA deficiency affects the ubiquitin-proteasome system function. DNA Repair (Amst) 2020; 94:102937. [PMID: 32693352 DOI: 10.1016/j.dnarep.2020.102937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/25/2020] [Accepted: 07/13/2020] [Indexed: 01/02/2023]
Abstract
Xeroderma pigmentosum complementation group A (XPA), is defective in xeroderma pigmentosum patients, causing pre-disposition to skin cancer and neurological abnormalities, which is not well understood. Here, we analyzed the XPA-deficient cells transcriptional profile under oxidative stress. The imbalance in of ubiquitin-proteasome system (UPS) gene expression was observed in XPA-deficient cells and the involvement of nuclear factor erythroid 2-related factor-2 (NFE2L2) was indicated. Co-immunoprecipitation assays showed the interaction between XPA, apurinic-apyrimidinic endonuclease 1 (APE1) and NFE2L2 proteins. Decreased NFE2L2 protein expression and proteasome activity was also observed in XPA-deficient cells. The data suggest the involvement of the growth arrest and DNA-damage-inducible beta (GADD45β) in NFE2L2 functions. Similar results were obtained in xpa-1 (RNAi) Caenorhabditis elegans suggesting the conservation of XPA and NFE2L2 interactions. In conclusion, stress response activation occurs in XPA-deficient cells under oxidative stress; however, these cells fail to activate the UPS cytoprotective response, which may contribute to XPA patient's phenotypes.
Collapse
|
14
|
Mutlu N, Sheidy DT, Hsu A, Jeong HS, Wozniak KJ, Kumar A. A Stress-Responsive Signaling Network Regulating Pseudohyphal Growth and Ribonucleoprotein Granule Abundance in Saccharomyces cerevisiae. Genetics 2019; 213:705-720. [PMID: 31455721 PMCID: PMC6781900 DOI: 10.1534/genetics.119.302538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/21/2019] [Indexed: 11/18/2022] Open
Abstract
The budding yeast Saccharomyces cerevisiae undergoes a stress-responsive transition to a pseudohyphal growth form in which cells elongate and remain connected in multicellular filaments. Pseudohyphal growth is regulated through conserved signaling networks that control cell growth and the response to glucose or nitrogen limitation in metazoans. These networks are incompletely understood, and our studies identify the TORC1- and PKA-regulated kinase Ksp1p as a key stress-responsive signaling effector in the yeast pseudohyphal growth response. The kinase-defective ksp1-K47D allele results in decreased pseudohyphal morphology at the cellular and colony level, indicating that Ksp1p kinase signaling is required for pseudohyphal filamentation. To determine the functional consequences of Ksp1p signaling, we implemented transcriptional profiling and quantitative phosphoproteomic analysis of ksp1-K47D on a global scale. Ksp1p kinase signaling maintains wild-type transcript levels of many pathways for amino acid synthesis and metabolism, relevant for the regulation of translation under conditions of nutrient stress. Proteins in stress-responsive ribonucleoprotein granules are regulated post-translationally by Ksp1p, and the Ksp1p-dependent phosphorylation sites S176 in eIF4G/Tif4631p and S436 in Pbp1p are required for wild-type levels of pseudohyphal growth and Protein Kinase A pathway activity. Pbp1p and Tif4631p localize in stress granules, and the ksp1 null mutant shows elevated abundance of Pbp1p puncta relative to wild-type. Collectively, the Ksp1p kinase signaling network integrates polarized pseudohyphal morphogenesis and translational regulation through the stress-responsive transcriptional control of pathways for amino acid metabolism and post-translational modification of translation factors affecting stress granule abundance.
Collapse
Affiliation(s)
- Nebibe Mutlu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Daniel T Sheidy
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Han Seol Jeong
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Katherine J Wozniak
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Anuj Kumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
- Program in Molecular and Cellular Biology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
15
|
Markov AV, Kel AE, Salomatina OV, Salakhutdinov NF, Zenkova MA, Logashenko EB. Deep insights into the response of human cervical carcinoma cells to a new cyano enone-bearing triterpenoid soloxolone methyl: a transcriptome analysis. Oncotarget 2019; 10:5267-5297. [PMID: 31523389 PMCID: PMC6731101 DOI: 10.18632/oncotarget.27085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
Semisynthetic triterpenoids, bearing cyano enone functionality in ring A, are considered now as novel promising anti-tumor agents. However, despite the large-scale studies, their effects on cervical carcinoma cells and, moreover, mechanisms underlying cell death activation by such compounds in this cell type have not been fully elucidated. In this work, we attempted to reconstitute the key pathways and master regulators involved in the response of human cervical carcinoma KB-3-1 cells to the novel glycyrrhetinic acid derivative soloxolone methyl (SM) by a transcriptomic approach. Functional annotation of differentially expressed genes, analysis of their cis- regulatory sequences and protein-protein interaction network clearly indicated that stress of endoplasmic reticulum (ER) is the central event triggered by SM in the cells. A range of key ER stress sensors and transcription factor AP-1 were identified as upstream transcriptional regulators, controlling the response of the cells to SM. Additionally, by using Gene Expression Omnibus data, we showed the ability of SM to modulate the expression of key genes involved in regulation of the high proliferative rate of cervical carcinoma cells. Further Connectivity Map analysis revealed similarity of SM's effects with known ER stress inducers thapsigargin and geldanamycin, targeting SERCA and Grp94, respectively. According to the molecular docking study, SM could snugly fit into the active sites of these proteins in the positions very close to that of both inhibitors. Taken together, our findings provide a basis for the better understanding of the intracellular processes in tumor cells switched on in response to cyano enone-bearing triterpenoids.
Collapse
Affiliation(s)
- Andrey V Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Alexander E Kel
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation.,geneXplain GmbH, Wolfenbüttel 38302, Germany
| | - Oksana V Salomatina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation.,N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Nariman F Salakhutdinov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Marina A Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Evgeniya B Logashenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| |
Collapse
|
16
|
Filamentation Regulatory Pathways Control Adhesion-Dependent Surface Responses in Yeast. Genetics 2019; 212:667-690. [PMID: 31053593 DOI: 10.1534/genetics.119.302004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/18/2019] [Indexed: 01/07/2023] Open
Abstract
Signaling pathways can regulate biological responses by the transcriptional regulation of target genes. In yeast, multiple signaling pathways control filamentous growth, a morphogenetic response that occurs in many species including fungal pathogens. Here, we examine the role of signaling pathways that control filamentous growth in regulating adhesion-dependent surface responses, including mat formation and colony patterning. Expression profiling and mutant phenotype analysis showed that the major pathways that regulate filamentous growth [filamentous growth MAPK (fMAPK), RAS, retrograde (RTG), RIM101, RPD3, ELP, SNF1, and PHO85] also regulated mat formation and colony patterning. The chromatin remodeling complex, SAGA, also regulated these responses. We also show that the RAS and RTG pathways coregulated a common set of target genes, and that SAGA regulated target genes known to be controlled by the fMAPK, RAS, and RTG pathways. Analysis of surface growth-specific targets identified genes that respond to low oxygen, high temperature, and desiccation stresses. We also explore the question of why cells make adhesive contacts in colonies. Cell adhesion contacts mediated by the coregulated target and adhesion molecule, Flo11p, deterred entry into colonies by macroscopic predators and impacted colony temperature regulation. The identification of new regulators (e.g., SAGA), and targets of surface growth in yeast may provide insights into fungal pathogenesis in settings where surface growth and adhesion contributes to virulence.
Collapse
|
17
|
Abstract
Filamentous growth is a fungal morphogenetic response that is critical for virulence in some fungal species. Many aspects of filamentous growth remain poorly understood. We have identified an aspect of filamentous growth in the budding yeast Saccharomyces cerevisiae and the human pathogen Candida albicans where cells behave collectively to invade surfaces in aggregates. These responses may reflect an extension of normal filamentous growth, as they share the same signaling pathways and effector processes. Aggregate responses may involve cooperation among individual cells, because aggregation was stimulated by cell adhesion molecules, secreted enzymes, and diffusible molecules that promote quorum sensing. Our study may provide insights into the genetic basis of collective cellular responses in fungi. The study may have ramifications in fungal pathogenesis, in situations where collective responses occur to promote virulence. Many fungal species, including pathogens, undergo a morphogenetic response called filamentous growth, where cells differentiate into a specialized cell type to promote nutrient foraging and surface colonization. Despite the fact that filamentous growth is required for virulence in some plant and animal pathogens, certain aspects of this behavior remain poorly understood. By examining filamentous growth in the budding yeast Saccharomyces cerevisiae and the opportunistic pathogen Candida albicans, we identify responses where cells undergo filamentous growth in groups of cells or aggregates. In S. cerevisiae, aggregate invasive growth was regulated by signaling pathways that control normal filamentous growth. These pathways promoted aggregation in part by fostering aspects of microbial cooperation. For example, aggregate invasive growth required cellular contacts mediated by the flocculin Flo11p, which was produced at higher levels in aggregates than cells undergoing regular invasive growth. Aggregate invasive growth was also stimulated by secreted enzymes, like invertase, which produce metabolites that are shared among cells. Aggregate invasive growth was also induced by alcohols that promote density-dependent filamentous growth in yeast. Aggregate invasive growth also required highly polarized cell morphologies, which may affect the packing or organization of cells. A directed selection experiment for aggregating phenotypes uncovered roles for the fMAPK and RAS pathways, which indicates that these pathways play a general role in regulating aggregate-based responses in yeast. Our study extends the range of responses controlled by filamentation regulatory pathways and has implications in understanding aspects of fungal biology that may be relevant to fungal pathogenesis. IMPORTANCE Filamentous growth is a fungal morphogenetic response that is critical for virulence in some fungal species. Many aspects of filamentous growth remain poorly understood. We have identified an aspect of filamentous growth in the budding yeast Saccharomyces cerevisiae and the human pathogen Candida albicans where cells behave collectively to invade surfaces in aggregates. These responses may reflect an extension of normal filamentous growth, as they share the same signaling pathways and effector processes. Aggregate responses may involve cooperation among individual cells, because aggregation was stimulated by cell adhesion molecules, secreted enzymes, and diffusible molecules that promote quorum sensing. Our study may provide insights into the genetic basis of collective cellular responses in fungi. The study may have ramifications in fungal pathogenesis, in situations where collective responses occur to promote virulence.
Collapse
|
18
|
Fang O, Hu X, Wang L, Jiang N, Yang J, Li B, Luo Z. Amn1 governs post-mitotic cell separation in Saccharomyces cerevisiae. PLoS Genet 2018; 14:e1007691. [PMID: 30273335 PMCID: PMC6181423 DOI: 10.1371/journal.pgen.1007691] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 10/11/2018] [Accepted: 09/12/2018] [Indexed: 11/18/2022] Open
Abstract
Post-mitotic cell separation is one of the most prominent events in the life cycle of eukaryotic cells, but the molecular underpinning of this fundamental biological process is far from being concluded and fully characterized. We use budding yeast Saccharomyces cerevisiae as a model and demonstrate AMN1 as a major gene underlying post-mitotic cell separation in a natural yeast strain, YL1C. Specifically, we define a novel 11-residue domain by which Amn1 binds to Ace2. Moreover, we demonstrate that Amn1 induces proteolysis of Ace2 through the ubiquitin proteasome system and in turn, down-regulates Ace2’s downstream target genes involved in hydrolysis of the primary septum, thus leading to inhibition of cell separation and clumping of haploid yeast cells. Using ChIP assays and site-specific mutation experiments, we show that Ste12 and the a1-α12 heterodimer are two direct regulators of AMN1. Specifically, a1-α2, a diploid-specific heterodimer, prevents Ste12 from inactivating AMN1 through binding to its promoter. This demonstrates how the Amn1-governed cell separation is highly cell type dependent. Finally, we show that AMN1368D from YL1C is a dominant allele in most strains of S. cerevisiae and evolutionarily conserved in both genic structure and phenotypic effect in two closely related yeast species, K. lactis and C. glabrata. Separation of mother and daughter cells after mitosis in eukaryotes enacts various functional and/or developmental needs and has significant medical and industrial implications. How this cellular behaviour is regulated is far from being concluded. We report here a novel Amn1 mediated post-mitotic cell separation in a budding yeast strain, YL1C and demonstrate that the post-mitotic cell separation can be regulated through a ubiquitin-conjugated protein degradation of Ace2 by Amn1. The Amn1-governed switch of cell separation is evolutionarily conserved and highly cell type dependent. These findings provide insights into the molecular mechanism of how post-mitotic cell separation is regulated in budding yeast, and data for translating into medical and industrial applications.
Collapse
Affiliation(s)
- Ou Fang
- Laboratory of Population and Quantitative Genetics, Institute of Biostatistics and Genetics, School of Life Sciences, Fudan University, Shanghai, China
- School of Biosciences, the University of Birmingham, Birmingham, United Kingdom
- Department of Evolution and Ecology, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaohua Hu
- Laboratory of Population and Quantitative Genetics, Institute of Biostatistics and Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Lin Wang
- Laboratory of Population and Quantitative Genetics, Institute of Biostatistics and Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Ning Jiang
- Laboratory of Population and Quantitative Genetics, Institute of Biostatistics and Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Jixuan Yang
- Laboratory of Population and Quantitative Genetics, Institute of Biostatistics and Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Bo Li
- Department of Evolution and Ecology, School of Life Sciences, Fudan University, Shanghai, China
| | - Zewei Luo
- Laboratory of Population and Quantitative Genetics, Institute of Biostatistics and Genetics, School of Life Sciences, Fudan University, Shanghai, China
- School of Biosciences, the University of Birmingham, Birmingham, United Kingdom
- * E-mail: ,
| |
Collapse
|
19
|
Impact of Fungal MAPK Pathway Targets on the Cell Wall. J Fungi (Basel) 2018; 4:jof4030093. [PMID: 30096860 PMCID: PMC6162559 DOI: 10.3390/jof4030093] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 12/14/2022] Open
Abstract
The fungal cell wall is an extracellular organelle that provides structure and protection to cells. The cell wall also influences the interactions of cells with each other and surfaces. The cell wall can be reorganized in response to changing environmental conditions and different types of stress. Signaling pathways control the remodeling of the cell wall through target proteins that are in many cases not well defined. The Mitogen Activated Protein Kinase pathway that controls filamentous growth in yeast (fMAPK) was required for normal growth in media containing the cell wall perturbing agent Calcofluor White (CFW). A mass spectrometry (MASS-SPEC) approach and analysis of expression profiling data identified cell wall proteins and modifying enzymes whose levels were influenced by the fMAPK pathway. These include Flo11p, Flo10p, Tip1p, Pry2p and the mannosyltransferase, Och1p. Cells lacking Flo11p or Och1p were sensitive to CFW. The identification of cell wall proteins controlled by a MAPK pathway may provide insights into how signaling pathways regulate the cell wall.
Collapse
|
20
|
Mukherjee S, Carignano A, Seelig G, Lee SI. Identifying progressive gene network perturbation from single-cell RNA-seq data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2018:5034-5040. [PMID: 30441472 DOI: 10.1109/embc.2018.8513444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Identifying the gene regulatory networks that control development or disease is one of the most important problems in biology. Here, we introduce a computational approach, called PIPER (ProgressIve network PERturbation), to identify the perturbed genes that drive differences in the gene regulatory network across different points in a biological progression. PIPER employs algorithms tailor-made for single cell RNA sequencing (scRNA-seq) data to jointly identify gene networks for multiple progressive conditions. It then performs differential network analysis along the identified gene networks to identify master regulators. We demonstrate that PIPER outperforms state-of-the-art alternative methods on simulated data and is able to predict known key regulators of differentiation on real scRNA-Seq datasets.
Collapse
|
21
|
Muetze T, Lynn DJ. Using the Contextual Hub Analysis Tool (CHAT) in Cytoscape to Identify Contextually Relevant Network Hubs. ACTA ACUST UNITED AC 2018; 59:8.24.1-8.24.13. [DOI: 10.1002/cpbi.35] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Tanja Muetze
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Health and Medical Research Institute North Terrace Adelaide Australia
| | - David J. Lynn
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Health and Medical Research Institute North Terrace Adelaide Australia
- School of Medicine, Flinders University Bedford Park Australia
| |
Collapse
|
22
|
Sarmiento‐Villamil JL, García‐Pedrajas NE, Baeza‐Montañez L, García‐Pedrajas MD. The APSES transcription factor Vst1 is a key regulator of development in microsclerotium- and resting mycelium-producing Verticillium species. MOLECULAR PLANT PATHOLOGY 2018; 19:59-76. [PMID: 27696683 PMCID: PMC6638171 DOI: 10.1111/mpp.12496] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 05/12/2023]
Abstract
Plant pathogens of the genus Verticillium pose a threat to many important crops worldwide. They are soil-borne fungi which invade the plant systemically, causing wilt symptoms. We functionally characterized the APSES family transcription factor Vst1 in two Verticillium species, V. dahliae and V. nonalfalfae, which produce microsclerotia and melanized hyphae as resistant structures, respectively. We found that, in V. dahliae Δvst1 strains, microsclerotium biogenesis stalled after an initial swelling of hyphal cells and cultures were never pigmented. In V. nonalfalfae Δvst1, melanized hyphae were also absent. These results suggest that Vst1 controls melanin biosynthesis independent of its role in morphogenesis. The absence of vst1 also had a great impact on sporulation in both species, affecting the generation of the characteristic verticillate conidiophore structure and sporulation rates in liquid medium. In contrast with these key roles in development, Vst1 activity was dispensable for virulence. We performed a microarray analysis comparing global transcription patterns of wild-type and Δvst1 in V. dahliae. G-protein/cyclic adenosine monophosphate (G-protein/cAMP) signalling and mitogen-activated protein kinase (MAPK) cascades are known to regulate fungal morphogenesis and virulence. The microarray analysis revealed a negative interaction of Vst1 with G-protein/cAMP signalling and a positive interaction with MAPK signalling. This analysis also identified Rho signalling as a potential regulator of morphogenesis in V. dahliae, positively interacting with Vst1. Furthermore, it exposed the association of secondary metabolism and development in this species, identifying Vst1 as a potential co-regulator of both processes. Characterization of the putative Vst1 targets identified in this study will aid in the dissection of specific aspects of development.
Collapse
Affiliation(s)
- Jorge L. Sarmiento‐Villamil
- Instituto de Hortofruticultura Subtropical y Mediterránea ‘La Mayora’ ‐ Universidad de Málaga ‐ Consejo Superior de Investigaciones Científicas (IHSM‐UMA‐CSIC), Estación Experimental ‘La Mayora’, 29750 Algarrobo‐CostaMálagaSpain
| | - Nicolás E. García‐Pedrajas
- Department of Computing and Numerical Analysis, C2 Building 3rd FloorCampus Universitario de RabanalesCórdoba14071Spain
| | - Lourdes Baeza‐Montañez
- Instituto de Hortofruticultura Subtropical y Mediterránea ‘La Mayora’ ‐ Universidad de Málaga ‐ Consejo Superior de Investigaciones Científicas (IHSM‐UMA‐CSIC), Estación Experimental ‘La Mayora’, 29750 Algarrobo‐CostaMálagaSpain
| | - María D. García‐Pedrajas
- Instituto de Hortofruticultura Subtropical y Mediterránea ‘La Mayora’ ‐ Universidad de Málaga ‐ Consejo Superior de Investigaciones Científicas (IHSM‐UMA‐CSIC), Estación Experimental ‘La Mayora’, 29750 Algarrobo‐CostaMálagaSpain
| |
Collapse
|
23
|
Yang B, Wittkopp PJ. Structure of the Transcriptional Regulatory Network Correlates with Regulatory Divergence in Drosophila. Mol Biol Evol 2017; 34:1352-1362. [PMID: 28333240 DOI: 10.1093/molbev/msx068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Transcriptional control of gene expression is regulated by biochemical interactions between cis-regulatory DNA sequences and trans-acting factors that form complex regulatory networks. Genetic changes affecting both cis- and trans-acting sequences in these networks have been shown to alter patterns of gene expression as well as higher-order organismal phenotypes. Here, we investigate how the structure of these regulatory networks relates to patterns of polymorphism and divergence in gene expression. To do this, we compared a transcriptional regulatory network inferred for Drosophila melanogaster to differences in gene regulation observed between two strains of D. melanogaster as well as between two pairs of closely related species: Drosophila sechellia and Drosophila simulans, and D. simulans and D. melanogaster. We found that the number of transcription factors predicted to directly regulate a gene ("in-degree") was negatively correlated with divergence in both gene expression (mRNA abundance) and cis-regulation. This observation suggests that the number of transcription factors directly regulating a gene's expression affects the conservation of cis-regulation and gene expression over evolutionary time. We also tested the hypothesis that transcription factors regulating more target genes (higher "out-degree") are less likely to evolve changes in their cis-regulation and expression (presumably due to increased pleiotropy), but found little support for this predicted relationship. Taken together, these data show how the architecture of regulatory networks can influence regulatory evolution.
Collapse
Affiliation(s)
- Bing Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Patricia J Wittkopp
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI.,Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
24
|
Abstract
Candida albicans is among the most prevalent fungal species of the human microbiota and asymptomatically colonizes healthy individuals. However, it is also an opportunistic pathogen that can cause severe, and often fatal, bloodstream infections. The medical impact of C. albicans typically depends on its ability to form biofilms, which are closely packed communities of cells that attach to surfaces, such as tissues and implanted medical devices. In this Review, we provide an overview of the processes involved in the formation of C. albicans biofilms and discuss the core transcriptional network that regulates biofilm development. We also consider some of the advantages that biofilms provide to C. albicans in comparison with planktonic growth and explore polymicrobial biofilms that are formed by C. albicans and certain bacterial species.
Collapse
|
25
|
Cromie GA, Tan Z, Hays M, Sirr A, Jeffery EW, Dudley AM. Transcriptional Profiling of Biofilm Regulators Identified by an Overexpression Screen in Saccharomyces cerevisiae. G3 (BETHESDA, MD.) 2017; 7:2845-2854. [PMID: 28673928 PMCID: PMC5555487 DOI: 10.1534/g3.117.042440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/27/2017] [Indexed: 12/25/2022]
Abstract
Biofilm formation by microorganisms is a major cause of recurring infections and removal of biofilms has proven to be extremely difficult given their inherent drug resistance . Understanding the biological processes that underlie biofilm formation is thus extremely important and could lead to the development of more effective drug therapies, resulting in better infection outcomes. Using the yeast Saccharomyces cerevisiae as a biofilm model, overexpression screens identified DIG1, SFL1, HEK2, TOS8, SAN1, and ROF1/YHR177W as regulators of biofilm formation. Subsequent RNA-seq analysis of biofilm and nonbiofilm-forming strains revealed that all of the overexpression strains, other than DIG1 and TOS8, were adopting a single differential expression profile, although induced to varying degrees. TOS8 adopted a separate profile, while the expression profile of DIG1 reflected the common pattern seen in most of the strains, plus substantial DIG1-specific expression changes. We interpret the existence of the common transcriptional pattern seen across multiple, unrelated overexpression strains as reflecting a transcriptional state, that the yeast cell can access through regulatory signaling mechanisms, allowing an adaptive morphological change between biofilm-forming and nonbiofilm states.
Collapse
Affiliation(s)
- Gareth A Cromie
- Pacific Northwest Research Institute, Seattle, Washington 98122
| | - Zhihao Tan
- Pacific Northwest Research Institute, Seattle, Washington 98122
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore 138648
| | - Michelle Hays
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington 98195
| | - Amy Sirr
- Pacific Northwest Research Institute, Seattle, Washington 98122
| | - Eric W Jeffery
- Pacific Northwest Research Institute, Seattle, Washington 98122
| | - Aimée M Dudley
- Pacific Northwest Research Institute, Seattle, Washington 98122
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington 98195
| |
Collapse
|
26
|
Hope EA, Amorosi CJ, Miller AW, Dang K, Heil CS, Dunham MJ. Experimental Evolution Reveals Favored Adaptive Routes to Cell Aggregation in Yeast. Genetics 2017; 206:1153-1167. [PMID: 28450459 PMCID: PMC5499169 DOI: 10.1534/genetics.116.198895] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/06/2017] [Indexed: 02/02/2023] Open
Abstract
Yeast flocculation is a community-building cell aggregation trait that is an important mechanism of stress resistance and a useful phenotype for brewers; however, it is also a nuisance in many industrial processes, in clinical settings, and in the laboratory. Chemostat-based evolution experiments are impaired by inadvertent selection for aggregation, which we observe in 35% of populations. These populations provide a testing ground for understanding the breadth of genetic mechanisms Saccharomyces cerevisiae uses to flocculate, and which of those mechanisms provide the biggest adaptive advantages. In this study, we employed experimental evolution as a tool to ask whether one or many routes to flocculation are favored, and to engineer a strain with reduced flocculation potential. Using a combination of whole genome sequencing and bulk segregant analysis, we identified causal mutations in 23 independent clones that had evolved cell aggregation during hundreds of generations of chemostat growth. In 12 of those clones, we identified a transposable element insertion in the promoter region of known flocculation gene FLO1, and, in an additional five clones, we recovered loss-of-function mutations in transcriptional repressor TUP1, which regulates FLO1 and other related genes. Other causal mutations were found in genes that have not been previously connected to flocculation. Evolving a flo1 deletion strain revealed that this single deletion reduces flocculation occurrences to 3%, and demonstrated the efficacy of using experimental evolution as a tool to identify and eliminate the primary adaptive routes for undesirable traits.
Collapse
Affiliation(s)
- Elyse A Hope
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| | - Clara J Amorosi
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| | - Aaron W Miller
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| | - Kolena Dang
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| | - Caiti Smukowski Heil
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| | - Maitreya J Dunham
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| |
Collapse
|
27
|
Joshua IM, Höfken T. From Lipid Homeostasis to Differentiation: Old and New Functions of the Zinc Cluster Proteins Ecm22, Upc2, Sut1 and Sut2. Int J Mol Sci 2017; 18:ijms18040772. [PMID: 28379181 PMCID: PMC5412356 DOI: 10.3390/ijms18040772] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/27/2017] [Accepted: 03/31/2017] [Indexed: 12/27/2022] Open
Abstract
Zinc cluster proteins are a large family of transcriptional regulators with a wide range of biological functions. The zinc cluster proteins Ecm22, Upc2, Sut1 and Sut2 have initially been identified as regulators of sterol import in the budding yeast Saccharomyces cerevisiae. These proteins also control adaptations to anaerobic growth, sterol biosynthesis as well as filamentation and mating. Orthologs of these zinc cluster proteins have been identified in several species of Candida. Upc2 plays a critical role in antifungal resistance in these important human fungal pathogens. Upc2 is therefore an interesting potential target for novel antifungals. In this review we discuss the functions, mode of actions and regulation of Ecm22, Upc2, Sut1 and Sut2 in budding yeast and Candida.
Collapse
Affiliation(s)
| | - Thomas Höfken
- Division of Biosciences, Brunel University London, Uxbridge UB8 3PH, UK.
| |
Collapse
|
28
|
González B, Mas A, Beltran G, Cullen PJ, Torija MJ. Role of Mitochondrial Retrograde Pathway in Regulating Ethanol-Inducible Filamentous Growth in Yeast. Front Physiol 2017; 8:148. [PMID: 28424625 PMCID: PMC5372830 DOI: 10.3389/fphys.2017.00148] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/24/2017] [Indexed: 12/17/2022] Open
Abstract
In yeast, ethanol is produced as a by-product of fermentation through glycolysis. Ethanol also stimulates a developmental foraging response called filamentous growth and is thought to act as a quorum-sensing molecule. Ethanol-inducible filamentous growth was examined in a small collection of wine/European strains, which validated ethanol as an inducer of filamentous growth. Wine strains also showed variability in their filamentation responses, which illustrates the striking phenotypic differences that can occur among individuals. Ethanol-inducible filamentous growth in Σ1278b strains was independent of several of the major filamentation regulatory pathways [including fMAPK, RAS-cAMP, Snf1, Rpd3(L), and Rim101] but required the mitochondrial retrograde (RTG) pathway, an inter-organellar signaling pathway that controls the nuclear response to defects in mitochondrial function. The RTG pathway regulated ethanol-dependent filamentous growth by maintaining flux through the TCA cycle. The ethanol-dependent invasive growth response required the polarisome and transcriptional induction of the cell adhesion molecule Flo11p. Our results validate established stimuli that trigger filamentous growth and show how stimuli can trigger highly specific responses among individuals. Our results also connect an inter-organellar pathway to a quorum sensing response in fungi.
Collapse
Affiliation(s)
- Beatriz González
- Departament de Bioquímica i Biotecnologia, Universitat Rovira i VirgiliTarragona, Spain
| | - Albert Mas
- Departament de Bioquímica i Biotecnologia, Universitat Rovira i VirgiliTarragona, Spain
| | - Gemma Beltran
- Departament de Bioquímica i Biotecnologia, Universitat Rovira i VirgiliTarragona, Spain
| | - Paul J Cullen
- Department of Biological Sciences, University at BuffaloBuffalo, NY, USA
| | - María Jesús Torija
- Departament de Bioquímica i Biotecnologia, Universitat Rovira i VirgiliTarragona, Spain
| |
Collapse
|
29
|
The Saccharomyces cerevisiae Cdk8 Mediator Represses AQY1 Transcription by Inhibiting Set1p-Dependent Histone Methylation. G3-GENES GENOMES GENETICS 2017; 7:1001-1010. [PMID: 28143948 PMCID: PMC5345701 DOI: 10.1534/g3.117.039586] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the budding yeast Saccharomyces cerevisiae, nutrient depletion induces massive transcriptional reprogramming that relies upon communication between transcription factors, post-translational histone modifications, and the RNA polymerase II holoenzyme complex. Histone H3Lys4 methylation (H3Lys4 me), regulated by the Set1p-containing COMPASS methyltransferase complex and Jhd2p demethylase, is one of the most well-studied histone modifications. We previously demonstrated that the RNA polymerase II mediator components cyclin C-Cdk8p inhibit locus-specific H3Lys4 3me independently of Jhd2p. Here, we identify loci subject to cyclin C- and Jhd2p-dependent histone H3Lys4 3me inhibition using chromatin immunoprecipitation (ChIP)-seq. We further characterized the independent and combined roles of cyclin C and Jhd2p in controlling H3Lys4 3me and transcription in response to fermentable and nonfermentable carbon at multiple loci. These experiments suggest that H3Lys4 3me alone is insufficient to induce transcription. Interestingly, we identified an unexpected role for cyclin C-Cdk8p in repressing AQY1 transcription, an aquaporin whose expression is normally induced during nutrient deprivation. These experiments, combined with previous work in other labs, support a two-step model in which cyclin C-Cdk8p mediate AQY1 transcriptional repression by stimulating transcription factor proteolysis and preventing Set1p recruitment to the AQY1 locus.
Collapse
|
30
|
Dissecting Gene Expression Changes Accompanying a Ploidy-Based Phenotypic Switch. G3-GENES GENOMES GENETICS 2017; 7:233-246. [PMID: 27836908 PMCID: PMC5217112 DOI: 10.1534/g3.116.036160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Aneuploidy, a state in which the chromosome number deviates from a multiple of the haploid count, significantly impacts human health. The phenotypic consequences of aneuploidy are believed to arise from gene expression changes associated with the altered copy number of genes on the aneuploid chromosomes. To dissect the mechanisms underlying altered gene expression in aneuploids, we used RNA-seq to measure transcript abundance in colonies of the haploid Saccharomyces cerevisiae strain F45 and two aneuploid derivatives harboring disomies of chromosomes XV and XVI. F45 colonies display complex “fluffy” morphologies, while the disomic colonies are smooth, resembling laboratory strains. Our two disomes displayed similar transcriptional profiles, a phenomenon not driven by their shared smooth colony morphology nor simply by their karyotype. Surprisingly, the environmental stress response (ESR) was induced in F45, relative to the two disomes. We also identified genes whose expression reflected a nonlinear interaction between the copy number of a transcriptional regulatory gene on chromosome XVI, DIG1, and the copy number of other chromosome XVI genes. DIG1 and the remaining chromosome XVI genes also demonstrated distinct contributions to the effect of the chromosome XVI disome on ESR gene expression. Expression changes in aneuploids appear to reflect a mixture of effects shared between different aneuploidies and effects unique to perturbing the copy number of particular chromosomes, including nonlinear copy number interactions between genes. The balance between these two phenomena is likely to be genotype- and environment-specific.
Collapse
|
31
|
Zinc Cluster Transcription Factors Alter Virulence in Candida albicans. Genetics 2016; 205:559-576. [PMID: 27932543 DOI: 10.1534/genetics.116.195024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/16/2016] [Indexed: 11/18/2022] Open
Abstract
Almost all humans are colonized with Candida albicans However, in immunocompromised individuals, this benign commensal organism becomes a serious, life-threatening pathogen. Here, we describe and analyze the regulatory networks that modulate innate responses in the host niches. We identified Zcf15 and Zcf29, two Zinc Cluster transcription Factors (ZCF) that are required for C. albicans virulence. Previous sequence analysis of clinical C. albicans isolates from immunocompromised patients indicates that both ZCF genes diverged during clonal evolution. Using in vivo animal models, ex vivo cell culture methods, and in vitro sensitivity assays, we demonstrate that knockout mutants of both ZCF15 and ZCF29 are hypersensitive to reactive oxygen species (ROS), suggesting they help neutralize the host-derived ROS produced by phagocytes, as well as establish a sustained infection in vivo Transcriptomic analysis of mutants under resting conditions where cells were not experiencing oxidative stress revealed a large network that control macro and micronutrient homeostasis, which likely contributes to overall pathogen fitness in host niches. Under oxidative stress, both transcription factors regulate a separate set of genes involved in detoxification of ROS and down-regulating ribosome biogenesis. ChIP-seq analysis, which reveals vastly different binding partners for each transcription factor (TF) before and after oxidative stress, further confirms these results. Furthermore, the absence of a dominant binding motif likely facilitates their mobility, and supports the notion that they represent a recent expansion of the ZCF family in the pathogenic Candida species. Our analyses provide a framework for understanding new aspects of the interface between C. albicans and host defense response, and extends our understanding of how complex cell behaviors are linked to the evolution of TFs.
Collapse
|
32
|
van Wijlick L, Swidergall M, Brandt P, Ernst JF. Candida albicansresponds to glycostructure damage by Ace2-mediated feedback regulation of Cek1 signaling. Mol Microbiol 2016; 102:827-849. [DOI: 10.1111/mmi.13494] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Lasse van Wijlick
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
- Manchot Graduate School Molecules of Infection, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| | - Marc Swidergall
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| | - Philipp Brandt
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| | - Joachim F. Ernst
- Department Biologie; Molekulare Mykologie, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
- Manchot Graduate School Molecules of Infection, Heinrich-Heine-Universität; 40225 Düsseldorf Germany
| |
Collapse
|
33
|
Keren L, Hausser J, Lotan-Pompan M, Vainberg Slutskin I, Alisar H, Kaminski S, Weinberger A, Alon U, Milo R, Segal E. Massively Parallel Interrogation of the Effects of Gene Expression Levels on Fitness. Cell 2016; 166:1282-1294.e18. [PMID: 27545349 DOI: 10.1016/j.cell.2016.07.024] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/05/2016] [Accepted: 07/18/2016] [Indexed: 02/02/2023]
Abstract
Data of gene expression levels across individuals, cell types, and disease states is expanding, yet our understanding of how expression levels impact phenotype is limited. Here, we present a massively parallel system for assaying the effect of gene expression levels on fitness in Saccharomyces cerevisiae by systematically altering the expression level of ∼100 genes at ∼100 distinct levels spanning a 500-fold range at high resolution. We show that the relationship between expression levels and growth is gene and environment specific and provides information on the function, stoichiometry, and interactions of genes. Wild-type expression levels in some conditions are not optimal for growth, and genes whose fitness is greatly affected by small changes in expression level tend to exhibit lower cell-to-cell variability in expression. Our study addresses a fundamental gap in understanding the functional significance of gene expression regulation and offers a framework for evaluating the phenotypic effects of expression variation.
Collapse
Affiliation(s)
- Leeat Keren
- Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel; Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Jean Hausser
- Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Maya Lotan-Pompan
- Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ilya Vainberg Slutskin
- Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hadas Alisar
- Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sivan Kaminski
- Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Adina Weinberger
- Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Uri Alon
- Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ron Milo
- Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eran Segal
- Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel; Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
34
|
Honigberg SM. Similar environments but diverse fates: Responses of budding yeast to nutrient deprivation. MICROBIAL CELL 2016; 3:302-328. [PMID: 27917388 PMCID: PMC5134742 DOI: 10.15698/mic2016.08.516] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diploid budding yeast (Saccharomyces cerevisiae) can adopt one
of several alternative differentiation fates in response to nutrient limitation,
and each of these fates provides distinct biological functions. When different
strain backgrounds are taken into account, these various fates occur in response
to similar environmental cues, are regulated by the same signal transduction
pathways, and share many of the same master regulators. I propose that the
relationships between fate choice, environmental cues and signaling pathways are
not Boolean, but involve graded levels of signals, pathway activation and
master-regulator activity. In the absence of large differences between
environmental cues, small differences in the concentration of cues may be
reinforced by cell-to-cell signals. These signals are particularly essential for
fate determination within communities, such as colonies and biofilms, where fate
choice varies dramatically from one region of the community to another. The lack
of Boolean relationships between cues, signaling pathways, master regulators and
cell fates may allow yeast communities to respond appropriately to the wide
range of environments they encounter in nature.
Collapse
Affiliation(s)
- Saul M Honigberg
- Division of Cell Biology and Biophysics, University of Missouri-Kansas City, 5007 Rockhill Rd, Kansas City MO 64110, USA
| |
Collapse
|
35
|
Muetze T, Goenawan IH, Wiencko HL, Bernal-Llinares M, Bryan K, Lynn DJ. Contextual Hub Analysis Tool (CHAT): A Cytoscape app for identifying contextually relevant hubs in biological networks. F1000Res 2016; 5:1745. [PMID: 27853512 PMCID: PMC5105880 DOI: 10.12688/f1000research.9118.2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/26/2016] [Indexed: 01/21/2023] Open
Abstract
Highly connected nodes (hubs) in biological networks are topologically important to the structure of the network and have also been shown to be preferentially associated with a range of phenotypes of interest. The relative importance of a hub node, however, can change depending on the biological context. Here, we report a Cytoscape app, the Contextual Hub Analysis Tool (CHAT), which enables users to easily construct and visualize a network of interactions from a gene or protein list of interest, integrate contextual information, such as gene expression or mass spectrometry data, and identify hub nodes that are more highly connected to contextual nodes (e.g. genes or proteins that are differentially expressed) than expected by chance. In a case study, we use CHAT to construct a network of genes that are differentially expressed in Dengue fever, a viral infection. CHAT was used to identify and compare contextual and degree-based hubs in this network. The top 20 degree-based hubs were enriched in pathways related to the cell cycle and cancer, which is likely due to the fact that proteins involved in these processes tend to be highly connected in general. In comparison, the top 20 contextual hubs were enriched in pathways commonly observed in a viral infection including pathways related to the immune response to viral infection. This analysis shows that such
contextual hubs are considerably more biologically relevant than degree-based hubs and that analyses which rely on the identification of hubs solely based on their connectivity may be biased towards nodes that are highly connected in general rather than in the specific context of interest. Availability: CHAT is available for Cytoscape 3.0+ and can be installed via the Cytoscape App Store (
http://apps.cytoscape.org/apps/chat).
Collapse
Affiliation(s)
- Tanja Muetze
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - Ivan H Goenawan
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - Heather L Wiencko
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Meath, Ireland
| | - Manuel Bernal-Llinares
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - Kenneth Bryan
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - David J Lynn
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia; School of Medicine, Flinders University, Bedford Park, Australia
| |
Collapse
|
36
|
Muetze T, Goenawan IH, Wiencko HL, Bernal-Llinares M, Bryan K, Lynn DJ. Contextual Hub Analysis Tool (CHAT): A Cytoscape app for identifying contextually relevant hubs in biological networks. F1000Res 2016; 5:1745. [PMID: 27853512 PMCID: PMC5105880 DOI: 10.12688/f1000research.9118.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/26/2016] [Indexed: 07/30/2023] Open
Abstract
UNLABELLED Highly connected nodes (hubs) in biological networks are topologically important to the structure of the network and have also been shown to be preferentially associated with a range of phenotypes of interest. The relative importance of a hub node, however, can change depending on the biological context. Here, we report a Cytoscape app, the Contextual Hub Analysis Tool (CHAT), which enables users to easily construct and visualize a network of interactions from a gene or protein list of interest, integrate contextual information, such as gene expression or mass spectrometry data, and identify hub nodes that are more highly connected to contextual nodes (e.g. genes or proteins that are differentially expressed) than expected by chance. In a case study, we use CHAT to construct a network of genes that are differentially expressed in Dengue fever, a viral infection. CHAT was used to identify and compare contextual and degree-based hubs in this network. The top 20 degree-based hubs were enriched in pathways related to the cell cycle and cancer, which is likely due to the fact that proteins involved in these processes tend to be highly connected in general. In comparison, the top 20 contextual hubs were enriched in pathways commonly observed in a viral infection including pathways related to the immune response to viral infection. This analysis shows that such contextual hubs are considerably more biologically relevant than degree-based hubs and that analyses which rely on the identification of hubs solely based on their connectivity may be biased towards nodes that are highly connected in general rather than in the specific context of interest. AVAILABILITY CHAT is available for Cytoscape 3.0+ and can be installed via the Cytoscape App Store ( http://apps.cytoscape.org/apps/chat).
Collapse
Affiliation(s)
- Tanja Muetze
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - Ivan H. Goenawan
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - Heather L. Wiencko
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Meath, Ireland
| | - Manuel Bernal-Llinares
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - Kenneth Bryan
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
| | - David J. Lynn
- EMBL Australia Biomedical Informatics Group, Infection & Immunity Theme, South Australian Medical and Health Research Institute, Adelaide, Australia
- School of Medicine, Flinders University, Bedford Park, Australia
| |
Collapse
|
37
|
Tollot M, Assmann D, Becker C, Altmüller J, Dutheil JY, Wegner CE, Kahmann R. The WOPR Protein Ros1 Is a Master Regulator of Sporogenesis and Late Effector Gene Expression in the Maize Pathogen Ustilago maydis. PLoS Pathog 2016; 12:e1005697. [PMID: 27332891 PMCID: PMC4917244 DOI: 10.1371/journal.ppat.1005697] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/20/2016] [Indexed: 12/31/2022] Open
Abstract
The biotrophic basidiomycete fungus Ustilago maydis causes smut disease in maize. Hallmarks of the disease are large tumors that develop on all aerial parts of the host in which dark pigmented teliospores are formed. We have identified a member of the WOPR family of transcription factors, Ros1, as major regulator of spore formation in U. maydis. ros1 expression is induced only late during infection and hence Ros1 is neither involved in plant colonization of dikaryotic fungal hyphae nor in plant tumor formation. However, during late stages of infection Ros1 is essential for fungal karyogamy, massive proliferation of diploid fungal cells and spore formation. Premature expression of ros1 revealed that Ros1 counteracts the b-dependent filamentation program and induces morphological alterations resembling the early steps of sporogenesis. Transcriptional profiling and ChIP-seq analyses uncovered that Ros1 remodels expression of about 30% of all U. maydis genes with 40% of these being direct targets. In total the expression of 80 transcription factor genes is controlled by Ros1. Four of the upregulated transcription factor genes were deleted and two of the mutants were affected in spore development. A large number of b-dependent genes were differentially regulated by Ros1, suggesting substantial changes in this regulatory cascade that controls filamentation and pathogenic development. Interestingly, 128 genes encoding secreted effectors involved in the establishment of biotrophic development were downregulated by Ros1 while a set of 70 “late effectors” was upregulated. These results indicate that Ros1 is a master regulator of late development in U. maydis and show that the biotrophic interaction during sporogenesis involves a drastic shift in expression of the fungal effectome including the downregulation of effectors that are essential during early stages of infection. The fungus Ustilago maydis is a pathogen of maize which induces tumor formation in the infected tissue. In these tumors huge amounts of fungal spores develop. As a biotrophic pathogen, U. maydis establishes itself in the plant with the help of a large number of secreted effector proteins. Many effector proteins are important for virulence because they counteract plant defense reactions. In this manuscript we have identified and characterized Ros1, a master regulator for the late stages of U. maydis development. This transcription factor is expressed late during infection and controls nuclear fusion, hyphal aggregation and late proliferation. ros1 mutants are still able to induce tumor formation but these are a dead end because they do not contain any spores. We show that Ros1 interferes with the early regulatory cascade controlled by a complex of two homeodomain proteins. In addition, Ros1 triggers a major switch in the effector repertoire, suggesting that different sets of effectors are needed for different stages of fungal development inside the plant.
Collapse
Affiliation(s)
- Marie Tollot
- Max Planck Institute for Terrestrial Microbiology, Department of Organismic Interactions, Marburg, Germany
| | - Daniela Assmann
- Max Planck Institute for Terrestrial Microbiology, Department of Organismic Interactions, Marburg, Germany
| | - Christian Becker
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Janine Altmüller
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Julien Y. Dutheil
- Max Planck Institute for Terrestrial Microbiology, Department of Organismic Interactions, Marburg, Germany
| | - Carl-Eric Wegner
- Max Planck Institute for Terrestrial Microbiology, Deparment of Biogeochemistry, Marburg, Germany
| | - Regine Kahmann
- Max Planck Institute for Terrestrial Microbiology, Department of Organismic Interactions, Marburg, Germany
- * E-mail:
| |
Collapse
|
38
|
Lai X, Wolkenhauer O, Vera J. Understanding microRNA-mediated gene regulatory networks through mathematical modelling. Nucleic Acids Res 2016; 44:6019-35. [PMID: 27317695 PMCID: PMC5291278 DOI: 10.1093/nar/gkw550] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
The discovery of microRNAs (miRNAs) has added a new player to the regulation of gene expression. With the increasing number of molecular species involved in gene regulatory networks, it is hard to obtain an intuitive understanding of network dynamics. Mathematical modelling can help dissecting the role of miRNAs in gene regulatory networks, and we shall here review the most recent developments that utilise different mathematical modelling approaches to provide quantitative insights into the function of miRNAs in the regulation of gene expression. Key miRNA regulation features that have been elucidated via modelling include: (i) the role of miRNA-mediated feedback and feedforward loops in fine-tuning of gene expression; (ii) the miRNA–target interaction properties determining the effectiveness of miRNA-mediated gene repression; and (iii) the competition for shared miRNAs leading to the cross-regulation of genes. However, there is still lack of mechanistic understanding of many other properties of miRNA regulation like unconventional miRNA–target interactions, miRNA regulation at different sub-cellular locations and functional miRNA variant, which will need future modelling efforts to deal with. This review provides an overview of recent developments and challenges in this field.
Collapse
Affiliation(s)
- Xin Lai
- Laboratory of Systems Tumour Immunology, Department of Dermatology, Erlangen University Hospital and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology & Bioinformatics, University of Rostock, Rostock, 18051, Germany Stellenbosch Institute for Advanced Study, Wallenberg Research Centre at Stellenbosch University, 7600, South Africa
| | - Julio Vera
- Laboratory of Systems Tumour Immunology, Department of Dermatology, Erlangen University Hospital and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, 91054, Germany
| |
Collapse
|
39
|
Abstract
In humans, microbial cells (including bacteria, archaea, and fungi) greatly outnumber host cells. Candida albicans is the most prevalent fungal species of the human microbiota; this species asymptomatically colonizes many areas of the body, particularly the gastrointestinal and genitourinary tracts of healthy individuals. Alterations in host immunity, stress, resident microbiota, and other factors can lead to C. albicans overgrowth, causing a wide range of infections, from superficial mucosal to hematogenously disseminated candidiasis. To date, most studies of C. albicans have been carried out in suspension cultures; however, the medical impact of C. albicans (like that of many other microorganisms) depends on its ability to thrive as a biofilm, a closely packed community of cells. Biofilms are notorious for forming on implanted medical devices, including catheters, pacemakers, dentures, and prosthetic joints, which provide a surface and sanctuary for biofilm growth. C. albicans biofilms are intrinsically resistant to conventional antifungal therapeutics, the host immune system, and other environmental perturbations, making biofilm-based infections a significant clinical challenge. Here, we review our current knowledge of biofilms formed by C. albicans and closely related fungal species.
Collapse
Affiliation(s)
- Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, California 95343;
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, California 94143;
| |
Collapse
|
40
|
Abstract
When transcription regulatory networks are compared among distantly related eukaryotes, a number of striking similarities are observed: a larger-than-expected number of genes, extensive overlapping connections, and an apparently high degree of functional redundancy. It is often assumed that the complexity of these networks represents optimized solutions, precisely sculpted by natural selection; their common features are often asserted to be adaptive. Here, we discuss support for an alternative hypothesis: the common structural features of transcription networks arise from evolutionary trajectories of "least resistance"--that is, the relative ease with which certain types of network structures are formed during their evolution.
Collapse
|
41
|
Woods K, Höfken T. The zinc cluster proteins Upc2 and Ecm22 promote filamentation in Saccharomyces cerevisiae by sterol biosynthesis-dependent and -independent pathways. Mol Microbiol 2015; 99:512-27. [PMID: 26448198 DOI: 10.1111/mmi.13244] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2015] [Indexed: 12/31/2022]
Abstract
The transition between a unicellular yeast form to multicellular filaments is crucial for budding yeast foraging and the pathogenesis of many fungal pathogens such as Candida albicans. Here, we examine the role of the related transcription factors Ecm22 and Upc2 in Saccharomyces cerevisiae filamentation. Overexpression of either ECM22 or UPC2 leads to increased filamentation, whereas cells lacking both ECM22 and UPC2 do not exhibit filamentous growth. Ecm22 and Upc2 positively control the expression of FHN1, NPR1, PRR2 and sterol biosynthesis genes. These genes all play a positive role in filamentous growth, and their expression is upregulated during filamentation in an Ecm22/Upc2-dependent manner. Furthermore, ergosterol content increases during filamentous growth. UPC2 expression also increases during filamentation and is inhibited by the transcription factors Sut1 and Sut2. The expression of SUT1 and SUT2 in turn is under negative control of the transcription factor Ste12. We suggest that during filamentation Ste12 becomes activated and reduces SUT1/SUT2 expression levels. This would result in increased UPC2 levels and as a consequence to transcriptional activation of FHN1, NPR1, PRR2 and sterol biosynthesis genes. Higher ergosterol levels in combination with the proteins Fhn1, Npr1 and Prr2 would then mediate the transition to filamentous growth.
Collapse
Affiliation(s)
- Kelly Woods
- Division of Biosciences, Brunel University London, Uxbridge, UK
| | - Thomas Höfken
- Division of Biosciences, Brunel University London, Uxbridge, UK
| |
Collapse
|
42
|
Large-Scale Analysis of Kinase Signaling in Yeast Pseudohyphal Development Identifies Regulation of Ribonucleoprotein Granules. PLoS Genet 2015; 11:e1005564. [PMID: 26447709 PMCID: PMC4598065 DOI: 10.1371/journal.pgen.1005564] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/10/2015] [Indexed: 01/10/2023] Open
Abstract
Yeast pseudohyphal filamentation is a stress-responsive growth transition relevant to processes required for virulence in pathogenic fungi. Pseudohyphal growth is controlled through a regulatory network encompassing conserved MAPK (Ste20p, Ste11p, Ste7p, Kss1p, and Fus3p), protein kinase A (Tpk2p), Elm1p, and Snf1p kinase pathways; however, the scope of these pathways is not fully understood. Here, we implemented quantitative phosphoproteomics to identify each of these signaling networks, generating a kinase-dead mutant in filamentous S. cerevisiae and surveying for differential phosphorylation. By this approach, we identified 439 phosphoproteins dependent upon pseudohyphal growth kinases. We report novel phosphorylation sites in 543 peptides, including phosphorylated residues in Ras2p and Flo8p required for wild-type filamentous growth. Phosphoproteins in these kinase signaling networks were enriched for ribonucleoprotein (RNP) granule components, and we observe co-localization of Kss1p, Fus3p, Ste20p, and Tpk2p with the RNP component Igo1p. These kinases localize in puncta with GFP-visualized mRNA, and KSS1 is required for wild-type levels of mRNA localization in RNPs. Kss1p pathway activity is reduced in lsm1Δ/Δ and pat1Δ/Δ strains, and these genes encoding P-body proteins are epistatic to STE7. The P-body protein Dhh1p is also required for hyphal development in Candida albicans. Collectively, this study presents a wealth of data identifying the yeast phosphoproteome in pseudohyphal growth and regulatory interrelationships between pseudohyphal growth kinases and RNPs. Eukaryotic cells affect precise changes in shape and growth in response to environmental and nutritional stress, enabling cell survival and wild-type function. The single-celled budding yeast provides a striking example, undergoing a set of changes under conditions of nitrogen or glucose limitation resulting in the formation of extended cellular chains or filaments. Related filamentous growth transitions are required for virulence in pathogenic fungi and have been studied extensively; however, the full scope of signaling underlying the filamentous growth transition remains to be determined. Here, we used a combination of genetics and proteomics to identify proteins that undergo phosphorylation dependent upon kinases required for filamentous growth. Within this protein set, we identified novel sites of phosphorylation in the yeast proteome and extensive phosphorylation of mRNA-protein complexes regulating mRNA decay and translation. The data indicate an interrelationship between filamentous growth and these ubiquitously conserved sites of RNA regulation: the RNA-protein complexes are required for the filamentous growth transition, and a well studied filamentous growth signaling kinase is required for wild-type numbers of RNA-protein complexes. This interdependence is previously unappreciated, highlighting an additional level of translational control underlying this complex growth transition.
Collapse
|
43
|
Intersecting transcription networks constrain gene regulatory evolution. Nature 2015; 523:361-5. [PMID: 26153861 PMCID: PMC4531262 DOI: 10.1038/nature14613] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/04/2015] [Indexed: 12/21/2022]
Abstract
Epistasis—the non-additive interactions between different genetic loci—constrains evolutionary pathways, blocking some and permitting others1–8. For biological networks such as transcription circuits, the nature of these constraints and their consequences are largely unknown. Here we describe the evolutionary pathways of a transcription network that controls the response to mating pheromone in yeasts9. A component of this network, the transcription regulator Ste12, has evolved two different modes of binding to a set of its target genes. In one group of species, Ste12 binds to specific DNA binding sites, while in another lineage it occupies DNA indirectly, relying on a second transcription regulator to recognize DNA. We show, through the construction of various possible evolutionary intermediates, that evolution of the direct mode of DNA binding was not directly accessible to the ancestor. Instead, it was contingent on a lineage-specific change to an overlapping transcription network with a different function, the specification of cell type. These results show that analyzing and predicting the evolution of cis-regulatory regions requires an understanding of their positions in overlapping networks, as this placement constrains the available evolutionary pathways.
Collapse
|
44
|
Comparative Analysis of Transmembrane Regulators of the Filamentous Growth Mitogen-Activated Protein Kinase Pathway Uncovers Functional and Regulatory Differences. EUKARYOTIC CELL 2015; 14:868-83. [PMID: 26116211 DOI: 10.1128/ec.00085-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/17/2015] [Indexed: 12/14/2022]
Abstract
Filamentous growth is a microbial differentiation response that involves the concerted action of multiple signaling pathways. In budding yeast, one pathway that regulates filamentous growth is a Cdc42p-dependent mitogen-activated protein kinase (MAPK) pathway. Several transmembrane (TM) proteins regulate the filamentous growth pathway, including the signaling mucin Msb2p, the tetraspan osmosensor Sho1p, and an adaptor Opy2p. The TM proteins were compared to identify common and unique features. Msb2p, Sho1p, and Opy2p associated by coimmunoprecipitation analysis but showed predominantly different localization patterns. The different localization patterns of the proteins resulted in part from different rates of turnover from the plasma membrane (PM). In particular, Msb2p (and Opy2p) were turned over rapidly compared to Sho1p. Msb2p signaled from the PM, and its turnover was a rate-limiting step in MAPK signaling. Genetic analysis identified unique phenotypes of cells overexpressing the TM proteins. Therefore, each TM regulator of the filamentous growth pathway has its own regulatory pattern and specific function in regulating filamentous growth. This specialization may be important for fine-tuning and potentially diversifying the filamentation response.
Collapse
|
45
|
Cheng C, Andrews E, Yan KK, Ung M, Wang D, Gerstein M. An approach for determining and measuring network hierarchy applied to comparing the phosphorylome and the regulome. Genome Biol 2015; 16:63. [PMID: 25880651 PMCID: PMC4404648 DOI: 10.1186/s13059-015-0624-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/10/2015] [Indexed: 12/16/2022] Open
Abstract
Many biological networks naturally form a hierarchy with a preponderance of downward information flow. In this study, we define a score to quantify the degree of hierarchy in a network and develop a simulated-annealing algorithm to maximize the hierarchical score globally over a network. We apply our algorithm to determine the hierarchical structure of the phosphorylome in detail and investigate the correlation between its hierarchy and kinase properties. We also compare it to the regulatory network, finding that the phosphorylome is more hierarchical than the regulome.
Collapse
Affiliation(s)
- Chao Cheng
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA. .,Institute for Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA. .,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA.
| | - Erik Andrews
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA.
| | - Koon-Kiu Yan
- Program in Computational Biology and Bioinformatics, Yale University, 260 Whitney Avenue, New Haven, CT, 06520, USA.
| | - Matthew Ung
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA.
| | - Daifeng Wang
- Program in Computational Biology and Bioinformatics, Yale University, 260 Whitney Avenue, New Haven, CT, 06520, USA.
| | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, 260 Whitney Avenue, New Haven, CT, 06520, USA. .,Department of Molecular Biophysics and Biochemistry, Yale University, 260 Whitney Avenue, New Haven, CT, 06520, USA. .,Department of Computer Science, Yale University, 260 Whitney Avenue, New Haven, CT, 06520, USA.
| |
Collapse
|
46
|
Li Z, Chen Y, Liu D, Zhao N, Cheng H, Ren H, Guo T, Niu H, Zhuang W, Wu J, Ying H. Involvement of glycolysis/gluconeogenesis and signaling regulatory pathways in Saccharomyces cerevisiae biofilms during fermentation. Front Microbiol 2015; 6:139. [PMID: 25755652 PMCID: PMC4337339 DOI: 10.3389/fmicb.2015.00139] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/05/2015] [Indexed: 11/16/2022] Open
Abstract
Compared to free (free-living) cells, biofilm cells show increased resistance and stability to high-pressure fermentation conditions, although the reasons underlying these phenomena remain unclear. Here, we investigated biofilm formation with immobilized Saccharomyces cerevisiae cells grown on fiber surfaces during the process of ethanol fermentation. The development of biofilm colonies was visualized by fluorescent labeling and confocal microscopy. RNA from yeast cells at three different biofilm development periods was extracted and sequenced by high-throughput sequencing. We quantitated gene expression differences between biofilm cells and free cells and found that 2098, 1556, and 927 genes were significantly differentially expressed, respectively. We also validated the expression of previously reported genes and identified novel genes and pathways under the control of this system. Statistical analysis revealed that biofilm genes show significant gene expression changes principally in the initial period of biofilm formation compared to later periods. Carbohydrate metabolism, amino acid metabolism, signal transduction, and oxidoreductase activity were needed for biofilm formation. In contrast to previous findings, we observed some differential expression performances of FLO family genes, indicating that cell aggregation in our immobilized fermentation system was possibly independent of flocculation. Cyclic AMP-protein kinase A and mitogen-activated protein kinase pathways regulated signal transduction pathways during yeast biofilm formation. We found that carbohydrate metabolism, especially glycolysis/gluconeogenesis, played a key role in the development of S. cerevisiae biofilms. This work provides an important dataset for future studies aimed at gaining insight into the regulatory mechanisms of immobilized cells in biofilms, as well as for optimizing bioprocessing applications with S. cerevisiae.
Collapse
Affiliation(s)
- Zhenjian Li
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China ; State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Yong Chen
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Dong Liu
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Nan Zhao
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Hao Cheng
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Hengfei Ren
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Ting Guo
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Huanqing Niu
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Wei Zhuang
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Jinglan Wu
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| | - Hanjie Ying
- National Engineering Research Center for Biotechnology, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China ; State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University Nanjing, China
| |
Collapse
|
47
|
Baeza-Montañez L, Gold SE, Espeso EA, García-Pedrajas MD. Conserved and Distinct Functions of the “Stunted” (StuA)-Homolog Ust1 During Cell Differentiation in the Corn Smut Fungus Ustilago maydis. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2015; 28:86-102. [PMID: 25208341 DOI: 10.1094/mpmi-07-14-0215-r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Ustilago maydis, causal agent of corn smut, can proliferate saprobically in a yeast form but its infectious filamentous form is an obligate parasite. Previously, we showed that Ust1, the first APSES (Asm1p, Phd1p, Sok2p, Efg1p, and StuAp) transcription factor functionally characterized in the phylum Basidiomycota, controlled morphogenesis and virulence in this species. Here, we further analyzed Ust1 function using multiple experimental approaches and determined that i) Ust1 activity was able to partially reverse stuA− conidiophore defects in Aspergillus nidulans; ii) in U. maydis, normal development and virulence were strongly dependent on precise induction or repression of Ust1 activity; iii) consistent with its role as a transcription factor regulating multiple processes, Ust1 accumulated in the nucleus at various stages of the life cycle; iv) however, it was undetectable at specific stages of pathogenic growth, indicating that Ust1 repression is part of normal development in planta; v) StuA response elements upstream of the ust1 open reading frame exhibited affinity for U. maydis DNA-binding proteins; vi) however, loss of regulated ust1 transcription had minor phenotypic effects; and vii) Ust1 was subject to post-translational phosphorylation but is not a target of cAMP signaling. Thus, the broad functional conservation between Ust1 and Ascomycota APSES proteins does not extend to the mechanisms regulating their activity.
Collapse
|
48
|
Sanchez P, Daniels KJ, Park YN, Soll DR. Generating a battery of monoclonal antibodies against native green fluorescent protein for immunostaining, FACS, IP, and ChIP using a unique adjuvant. Monoclon Antib Immunodiagn Immunother 2014; 33:80-8. [PMID: 24746148 DOI: 10.1089/mab.2013.0089] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Using a unique, nontoxic adjuvant compound of poly(I:C) and anti-CD40 MAb, a battery of eight mouse monoclonal antibodies was generated against native green fluorescent protein. All were effective to varying degrees for immunostaining paraformaldehyde-fixed cells, six for staining sections of paraffin-embedded tissue, all to varying degrees in fluorescent-activated cell sorting, five for immunoprecipitation, and seven for chromatin immunoprecipitation. None worked in denaturing Western blots since the target was the native GFP protein. Both the hybridomas and antibodies are available at cost through DSHB, a non-profit National Resource created by the National Institutes of Health.
Collapse
Affiliation(s)
- Philip Sanchez
- Developmental Studies Hybridoma Bank, The University of Iowa , Iowa City, Iowa
| | | | | | | |
Collapse
|
49
|
Fine-tuning of histone H3 Lys4 methylation during pseudohyphal differentiation by the CDK submodule of RNA polymerase II. Genetics 2014; 199:435-53. [PMID: 25467068 DOI: 10.1534/genetics.114.172841] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Transcriptional regulation is dependent upon the interactions between the RNA pol II holoenzyme complex and chromatin. RNA pol II is part of a highly conserved multiprotein complex that includes the core mediator and CDK8 subcomplex. In Saccharomyces cerevisiae, the CDK8 subcomplex, composed of Ssn2p, Ssn3p, Ssn8p, and Srb8p, is thought to play important roles in mediating transcriptional control of stress-responsive genes. Also central to transcriptional control are histone post-translational modifications. Lysine methylation, dynamically balanced by lysine methyltransferases and demethylases, has been intensively studied, uncovering significant functions in transcriptional control. A key question remains in understanding how these enzymes are targeted during stress response. To determine the relationship between lysine methylation, the CDK8 complex, and transcriptional control, we performed phenotype analyses of yeast lacking known lysine methyltransferases or demethylases in isolation or in tandem with SSN8 deletions. We show that the RNA pol II CDK8 submodule components SSN8/SSN3 and the histone demethylase JHD2 are required to inhibit pseudohyphal growth-a differentiation pathway induced during nutrient limitation-under rich conditions. Yeast lacking both SSN8 and JHD2 constitutively express FLO11, a major regulator of pseudohyphal growth. Interestingly, deleting known FLO11 activators including FLO8, MSS11, MFG1, TEC1, SNF1, KSS1, and GCN4 results in a range of phenotypic suppression. Using chromatin immunoprecipitation, we found that SSN8 inhibits H3 Lys4 trimethylation independently of JHD2 at the FLO11 locus, suggesting that H3 Lys4 hypermethylation is locking FLO11 into a transcriptionally active state. These studies implicate the CDK8 subcomplex in fine-tuning H3 Lys4 methylation levels during pseudohyphal differentiation.
Collapse
|
50
|
Li B, Gaudinier A, Tang M, Taylor-Teeples M, Nham NT, Ghaffari C, Benson DS, Steinmann M, Gray JA, Brady SM, Kliebenstein DJ. Promoter-based integration in plant defense regulation. PLANT PHYSIOLOGY 2014; 166:1803-20. [PMID: 25352272 PMCID: PMC4256871 DOI: 10.1104/pp.114.248716] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/28/2014] [Indexed: 05/18/2023]
Abstract
A key unanswered question in plant biology is how a plant regulates metabolism to maximize performance across an array of biotic and abiotic environmental stresses. In this study, we addressed the potential breadth of transcriptional regulation that can alter accumulation of the defensive glucosinolate metabolites in Arabidopsis (Arabidopsis thaliana). A systematic yeast one-hybrid study was used to identify hundreds of unique potential regulatory interactions with a nearly complete complement of 21 promoters for the aliphatic glucosinolate pathway. Conducting high-throughput phenotypic validation, we showed that >75% of tested transcription factor (TF) mutants significantly altered the accumulation of the defensive glucosinolates. These glucosinolate phenotypes were conditional upon the environment and tissue type, suggesting that these TFs may allow the plant to tune its defenses to the local environment. Furthermore, the pattern of TF/promoter interactions could partially explain mutant phenotypes. This work shows that defense chemistry within Arabidopsis has a highly intricate transcriptional regulatory system that may allow for the optimization of defense metabolite accumulation across a broad array of environments.
Collapse
Affiliation(s)
- Baohua Li
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Allison Gaudinier
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Michelle Tang
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Mallorie Taylor-Teeples
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Ngoc T Nham
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Cyrus Ghaffari
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Darik Scott Benson
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Margaret Steinmann
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Jennifer A Gray
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Siobhan M Brady
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| | - Daniel J Kliebenstein
- Departments of Plant Sciences (B.L., M.T., N.T.N. C.G., D.S.B., M.S., J.A.G., D.J.K.) and Plant Biology (A.G., M.T., M.T.-T., J.A.G., S.M.B.) and Genome Center (A.G., M.T., M.T.-T., J.A.G., S.M.B.), University of California, Davis, California 95616; andDynaMo Center of Excellence, University of Copenhagen, DK-1871 Frederiksberg C, Denmark (D.J.K.)
| |
Collapse
|