CSSB Centre for Structural Systems Biology
Suche
Home Research Research Groups Barisch Group

Barisch Group

Host-Microbe Interactome

Prof. Dr. Caroline Barisch

Group Leader

+49 40 8998 87620
caroline.barisch@cssb-hamburg.de

Home Institute

Research Center Borstel
fz-borstel.de/index.php/en/

Our research focus is to decipher the molecular mechanisms by which pathogenic mycobacteria remodel, acquire, import and utilise lipids from their host to support infection.

Research Projects

MISSION

Lipid Dynamics during Mycobacteria Infection

Tuberculosis (Tb) is caused by Mycobacterium tuberculosis and remains one of the deadliest infectious diseases. The World Health Organization (WHO) estimates that in 2021, Tb killed 1.6 million people emphasizing the importance to develop new drugs, vaccines and diagnostic tools to reduce this burden in the future.

M. tuberculosis employs multiple strategies to survive intracellularly. One of its most striking adaptations is its ability to utilize host lipids such as fatty acids and sterols to: (i) generate energy, (ii) build its characteristic lipid-rich cell wall and (iii) produce storage lipids during infection. To be constantly in a fatty acid-rich environment, the pathogen actively contributes to generate the “foamy” phenotype in host macrophages, for which the accumulation of host lipid droplets (LDs) is characteristic. 

Using the Dictyostelium discoideum/M. marinum infection system, we found that mycobacteria access host LDs to build up their own lipid storage organelles and exploit ER-derived phospholipids when LDs are lacking (Barisch et al., 2015; Barisch & Soldati, 2017). Moreover, we observed that mycobacteria that escaped from the Mycobacterium-containing vacuole (MCV) into the cytosol recruit LD-derived enzymes and regulatory proteins on their hydrophobic surface.

KEY INTEREST

The Barisch lab aims to unravel the molecular mechanisms by which pathogenic mycobacteria acquire lipids from their host to support chronic infection. Combining the application of functionalized lipid probes with mass spectrometry-based lipidomics and advanced microscopy techniques, the group analyses metabolic lipid flows between mycobacteria and their host at the subcellular and ultrastructural level.

PROJECTS

Balancing Act: How a pertubation in fatty acid homeostasis impacts on vacuole escape of mycobacteria

To import fatty acids from their environment, mycobacteria are equipped with sophisticated transport machineries. However, the mechanism by which fatty acids are esterified with coenzyme A (“fatty acid activation”), an essential step for their further turnover, remains elusive. ​This project aims to characterize the function of fatty acid-activating enzymes in lipid synthesis and vacuolar escape of mycobacteria using the D. discoideum/M. marinum system.

​​To characterize fatty acid flows and metabolism in host and bacteria mutants depleted in fatty acid-activating enzymes, a protocol that combines the use of bifunctional FA probes with expansion microscopy and lipidomics is established. - This project is part of the SPP2225.

Functional impact of lipid logistics during mycobacteria infection

This project aims to identify lipid metabolic pathways that are hijacked by intracellular mycobacteria to exploit lipids from the host. To monitor alterations in lipid levels, we are establishing mass spectrometry lipidomics and thin layer chromatography for the D. discoideum/M. marinum system. In the future, we will determine the consequences of blocking specific lipid supply routes on various stages of the mycobacterial infection course. Collectively, these efforts may uncover novel therapeutic targets to fight mycobacteria infection.

Induction of membrane contact sites during mycobacteria infection

Various intracellular pathogens, including M. tuberculosis, damage the membrane of their vacuoles to impair fundamental innate immune functions and to trigger their translocation into the host cytosol. The host counteracts membrane damage by recruiting membrane repair machineries to retain the pathogen inside the vacuole. ​

Using advanced imaging approaches, the lab investigates the role of ER-dependent membrane repair and other repair machineries during mycobacteria infection. For example, to investigate the formation of membrane contact sites between the ER and the Mycobacterium-containing vacuole (MCV), we employ advanced imaging techniques. Specifically, we utilize several 3D-CLEM approaches that include high-pressure freezing and TEM-tomography (as described in Franzkoch and Anand et al., 2023, BioRxiv) as well as serial block-face SEM (Anand et al., 2023, BioRxiv). This, together with spinning disc live cell imaging and flow cytometry, uncovered that ER-dependent repair constitutes a host defense mechanism against intracellular pathogens such as M. tuberculosis (Anand et al., 2023, BioRxiv).  - This project is part of the SFB1557 @Uni Osnabrück.

Research Team

8

Group Leader

Prof. Dr. Caroline Barisch
Phone:+49 40 8998 87620

E-Mail

Postdoctoral Scientist

Aby Anand
Phone:+49 40 8998 87621

E-Mail

Research Technician

Béla Boumazy
Phone:+49 40 8998 87621

E-Mail

PhD Student

Paulo Glatz
Phone:+49 40 8998 87621

E-Mail

Bachelor Student

Michelle Hacker
Phone:+49 40 8998 87621

E-Mail

PhD Student

Sylvana Victoria Hüttel
Phone:+49 40 8998 87621

E-Mail

PhD Student

Anna-Carina Mazur
Phone:+49 40 8998 87621

E-Mail

Master Student

Danica Müller


Publications

2023

Anand A, Mazur AC, Rosell-Arevalo P, Franzkoch R, Breitsprecher L, Listian SA, Hüttel SV, Müller D, Schäfer DG, Vormittag S, Hilbi H, Maniak M, Gutierrez MG, Barisch C (2023) ER-dependent membrane repair of mycobacteria-induced vacuole damage. mBio. e0094323. doi: 10.1128/mbio.00943-23.

Listian SA, Kol M, Ufelmann E, Eising S, Fröhlich F, Walter S, Holthuis JCM, Barisch C (2023). Complex sphingolipid profiling and identification of an inositol phosphorylceramide synthase in Dictyostelium discoideum. BioRxiv.

*Franzkock R, *Anand A, Psathaki OE, Barisch C (2023). Resolving exit strategies of mycobacteria by combining high-pressure freezing with 3D-correlative light and electron microscopy. BioRxiv. *authors contributed equally

Barisch C, Holthuis JCM, Cosentino K (2023). Membrane damage and repair: A thin line between life and death. Biol. Chem. REVIEW.

Vormittag S, Hüsler D, Haneburger I, Kroniger T, Anand A, Prantl M, Barisch C, Maaß S, Becher D, Letourneur F, Hilbi H (2023). Legionella- and host-driven lipid flux at LCV-ER membrane contact sites promotes vacuole remodeling.  EMBO Rep.

2022

Foulon M, Listian S A, Soldati T, Barisch C (2022). Chapter 6. Conserved mechanisms drive host lipid access, import and utilisation in Mycobacterium tuberculosis and M. marinum. In Developments in Microbiology, Biology of Mycobacterial Lipids, Academic Press. Edited by Fatima Z, Canaan S. REVIEW.

2021

Hanna N, Koliwer-Brandl H, Lefrançois L H, Kalinina V, Cardenal-Muñoz E, Appiah J, Leuba F, Hilbi H, Soldati T, Barisch C (2021). Zn2+ intoxication of Mycobacterium marinum during Dictyostelium discoideum infection is counteracted by induction of the pathogen Zn2+ exporter CtpC. mBio.

Niekamp P, Guzman G, Leier HC, Rashidfarrokhi A, Richina V, Pott F, Barisch C, Holthuis JCM, Tafesse FG (2021). Sphingomyelin biosynthesis is essential for phagocytic signaling during Mycobacterium tuberculosis host cell entry. mBio.

2020

Knobloch P, Koliwer-Brandl H, Arnold FM, Hanna N, Gonda I, Adenau S, Personnic N, Barisch C, Seeger MA, Soldati T & Hilbi H (2020). Mycobacterium marinum produces distinct mycobactin and carboxymycobactin siderophores to promote growth in broth and phagocytes. Cell Microbiol.

2019

Luscher A, Fröhlich F, Barisch C, Littlewood C, Metcalfe J, Leuba F, Palma A, Pirruccello M, Cesareni G, Stagi M, Walther TC, Soldati T, De Camilli P and Swan LE (2019). Lowe syndrome-linked endocytic adaptors direct membrane cycling kinetics with OCRL in Dictyostelium discoideum. Mol Biol Cell.

Koliwer-Brandl H, Knobloch P, Barisch C, Welin A, Hanna N, Soldati T and Hilbi H (2019). Distinct Mycobacterium marinum phosphatases determine pathogen vacuole phosphoinositide pattern, phagosome maturation, and escape to the cytosol. Cell Microbiol.

López-Jiménez AT, Cardenal-Muñoz E, Leuba F, Gerstenmaier L, Barisch C, Hagedorn M, King JS and Soldati T (2019). The ESCRT and autophagy machineries cooperate to repair ESX-1-dependent damage at the Mycobacterium-containing vacuole but have opposite impact on containing the infection. PLoS Pathog.

2018

Barisch C, Kalinina V, Lefrançois LH, Appiah J, López-Jiménez AT and Soldati T (2018). Localization of all four ZnT zinc transporters in Dictyostelium and impact of ZntA and ZntB knockout on bacteria killing. J Cell Sci.

Sattler N, Bosmani C, Barisch C, Guého A, Gopaldass N, Dias M, Leuba F, Bruckert F, Cosson P and Soldati T (2018). Functions of the Dictyostelium LIMP-2 and CD36 homologues in bacteria uptake, phagolysosome biogenesis and host cell defence. J Cell Sci.

Watkins RA, Andrews A, Wynn C, Barisch C, King JS and Johnston SA (2018). Cryptococcus neoformans Escape From Dictyostelium Amoeba by Both WASH-Mediated Constitutive Exocytosis and Vomocytosis. Front Cell Infect Microbiol.

Cardenal-Muñoz E, Barisch C, Lefrançois LH, López-Jiménez AT and Soldati T (2018). When Dicty Met Myco, a (Not So) Romantic Story about One Amoeba and Its Intracellular Pathogen. Front Cell Infect Microbiol. REVIEW.

Dunn JD, Bosmani C, Barisch C, Raykov L, Lefrançois LH, Cardenal-Muñoz E, López-Jiménez AT and Soldati T (2018). Eat Prey, Live: Dictyostelium discoideum As a Model for Cell-Autonomous Defenses. Front Immunol. REVIEW.

2017

Barisch C, Soldati T (2017). Breaking fat! How mycobacteria and other intracellular pathogens manipulate host lipid droplets. Biochimie. REVIEW.

Barisch C, Soldati T (2017). Mycobacterium marinum Degrades Both Triacylglycerols and Phospholipids from Its Dictyostelium Host to Synthesise Its Own Triacylglycerols and Generate Lipid Inclusions. PLoS Pathog.
Video clip (in French) on Radio Television Suisse: https://avisdexperts.ch/experts/caroline_barisch

2015

Barisch C, López-Jiménez AT and Soldati T (2015). Live imaging of Mycobacterium marinum infection in Dictyostelium discoideum. Methods Mol Biol. BOOK CHAPTER.

Barisch C, Paschke P, Hagedorn M, Maniak M and Soldati T (2015). Lipid droplet dynamics at early stages of Mycobacterium marinum infection in Dictyostelium. Cell Microbiol.

2013

Du X, Barisch C, Paschke P, Herrfurth C, Bertinetti O, Pawolleck N, Otto H, Rühling H, Feussner I, Herberg FW and Maniak M (2013). Dictyostelium lipid droplets host novel proteins. Eukaryot Cell.