A04: Platelet aggregation and activity in steatosis-HCC

Overweight and metabolic syndrome have reached pandemic dimensions in industrialized countries and are on the rise in developed countries (e.g. India, China). Clinically these pathologies can manifest in non-alcoholic fatty liver disease (NAFLD), the most frequent liver disease world-wide. A significant number of NAFLD patients develop non-alcoholic steatohepatitis (NASH), fibrosis, and hepatocellular carcinoma (HCC). The mechanisms triggering NASH and its transition to HCC remain elusive. Consequently, efficacious therapies are lacking. We have established a mouse model of NASH-driven HCC recapitulating human pathophysiology in the context of metabolic syndrome. We demonstrated that CD8+ and NKT cells become activated during metabolic syndrome, interact with hepatocytes, alter hepatic lipid metabolism, and cause NASH and HCC, in part through intrahepatic NF-κb signaling. Identical CD8+ and NKT cell activation profiles were found in human NASH livers underlining the clinical relevance of our model. Recent publications indicate a role of platelets in promoting T cell-triggered hepatitis in the context of virus infections. Our preliminary data demonstrate that blocking of platelet activation and aggregation using several clinically used therapeutic inhibitors prevents intrahepatic inflammation, NASH, and HCC development. However, the underlying mechanisms remain elusive. We will use distinct knock-out mouse models (e.g. vWF-/-; GPIb-/-; GPIIb-/-, GPIIa-/-; Nbeal2-/-; GpVI-/-) to define the exact molecular mechanisms by which platelet function or activity promote hepatocarcinogenesis in the context of NASH. Moreover, using proteomic or flow cytometric analyses of platelets we will further identify and test novel targets of functional, diagnostic, and therapeutic value for NASH driven HCC in mice and humans. Further, we will also analyze the hepatic metabolic changes and the environmental alterations and their direct consequences on tumor cell growth imposed by altered platelet function, with the final goal to genetically dissect the sole impact of metabolic genes and environmental factors (e.g. activation/ polarization of immune cells) on NASH to HCC transition. Data will be corroborated in human patient and serum samples. The long term aim of this proposal is to implement identified candidates in the frame of targeted treatment to suppress platelet aggregation/activation (e.g. using antibodies; small molecules) in order to block NASH-triggered hepatocarcinogenesis without imposing the adverse effects of classical anti-platelet aggregation therapy.

Publications

Peer-reviewed publications and books
• Rappez L&, Stadler M&, Triana S, Gathungu RM, Ovchinnikova K, Phapale P, Heikenwalder M*, Aelxandrov T* (2021). SpaceM reveals metabolic states of single cells. Nat Methods 18:799-805. &co-first authors, *co-last, co-corresponding author.
• Deczkowska A, David E, Ramadori P, Pfister D, Safran M, At The B, Giladi A, Jaitin DA, Barboy O, Cohen M, Yofe I, Gur C, Shlomi-Loubaton S, Henri S, Suhail Y, Qiu M, Kam S, Hermon H, Lahat E, Ben-Yakov G, Cohen-Ezra O, Davidov Y, Likhter M, Goitein D, Roth S, Weber A, Malissen B, Weiner A*, Ben-Ari Z*, Heikenwalder M*, Elinav E*, Amit I* (2021). XCR1+ type 1 conventional dendritic cells drive liver pathology in Non-Alcoholic Steatohepatitis. Nat Med 27:1043-1054. *co-last, co-corresponding author.
• Pfister D, Núñez NG, Pinyol R, Govaere O, Pinter M, Szydlowska M, Gupta R, Qiu M, Deczkowska A, Weiner A, Müller F, Sinha A, Friebel E, Engleitner T, Lenggenhager D, Moncsek A, Heide D, Stirm K, Kosla J, Kotsiliti E, Leone V, Dudek M, Yousuf S, Inverso D, Singh I, Teijeiro A, Castet F, Montironi C, Haber PK, Tiniakos D, Bedossa P, Cockell S, Younes R, Vacca M, Marra F, Schattenberg JM, Allison M, Bugianesi E, Ratziu V, Pressiani T, D'Alessio A, Personeni N, Rimassa L, Daly AK, Scheiner B, Pomej K, Kirstein MM, Vogel A, Peck-Radosavljevic M, Hucke F, Finkelmeier F, Waidmann O, Trojan J, Schulze K, Wege H, Koch S, Weinmann A, Bueter M, Rössler F, Siebenhüner A, Dosso S, Mallm JP, Umansky V, Jugold M, Luedde T, Schietinger A, Schirmacher P, Emu B, Augustin HG, Billeter A, Müller-Stich B, Kikuchi H, Duda DG, Kütting F, Waldschmidt DT, Ebert MP, Rahbari N, Mei HE, Schulz AR, Ringelhan M, Malek N, Spahn S, Bitzer M, Ruiz de Galarreta M, Lujambio A, Dufour JF, Marron TU, Kaseb A, Kudo M, Huang YH, Djouder N, Wolter K, Zender L, Marche PN, Decaens T, Pinato DJ, Rad R, Mertens JC, Weber A, Unger K, Meissner F, Roth S, Jilkova ZM, Claassen M, Anstee QM, Amit I, Knolle P, Becher B, Llovet JM*, Heikenwalder M* (2021). NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 592:450-456. *equal contribution.
• Dudek M, Pfister D, Donakonda S, Filpe P, Schneider A, Laschinger M, Hartmann D, Hüser N, Meiser P, Bayerl F, Inverso D, Wigger J, Sebode M, Öllinger R, Rad R, Hegenbarth S, Anton M, Guillot A, Bowman A, Heide D, Müller F, Ramadori P, Leone V, Garcia-Caceres C, Gruber T, Seifert G, Kabat AM, Malm JP, Reider S, Effenberger M, Roth S, Billeter AT, Müller-Stich B, Pearce EJ, Koch-Nolte F, Käser R, Tilg H, Thimme R, Böttler T, Tacke F, Dufour JF, Haller D, Murray PJ, Heeren R, Zehn D, Böttcher JP, Heikenwälder M, Knolle PA (2021). Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH. Nature 592:444-449.
• Namineni S, O‘Connor T, Faure-Dupuy S, Johansen P, Riedl T, Liu K, Xu H, Singh I, Shinde P, Li F, Pandyra A, Sharma P, Ringelhan M, Muschaweckh A, Borst K, Blank P, Lampl S, Durantel D, Farhat R, Weber A, Lenggenhager D, Kündig TM, Staeheli P, Protzer U, Wohlleber D, Holzmann B, Binder M, Breuhahn K, Assmus LM, Nattermann J, Abdullah Z, Rolland M, Dejardin E, Lang PA, Lang KS, Karin M, Lucifora J, Kalinke U, Knolle PA, Heikenwalder M (2020) A dual role for hepatocyte-intrinsic canonical NF-κB signaling in virus control. J Hepatol 72:960-975.
• Ramadori P, Klag T, Malek NP, Heikenwalder M (2019) Platelets in chronic liver disease, from bench to bedside. JHEP Rep 1:448-459.
• Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M (2019) From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol 16:411-428.
• Malehmir M, Pfister D, Gallage S, Szydlowska M, Inverso D, Kotsiliti E, Leone V, Peiseler M, Surewaard BGJ, Rath D, Ali A, Wolf MJ, Drescher H, Healy ME, Dauch D, Kroy D, Krenkel O, Kohlhepp M, Engleitner T, Olkus A, Sijmonsma T, Volz J, Deppermann C, Stegner D, Helbling P, Nombela-Arrieta C, Rafiei A, Hinterleitner M, Rall M, Baku F, Borst O, Wilson CL, Leslie J, O'Connor T, Weston CJ, Withers DJ, Ware J, Mann DA, Augustin HG, Vegiopoulos A, Milsom MD, Rose AJ, Lalor PF, Llovet JM, Pinyol R, Tacke F, Rad R, Matter M, Djouder N, Kubes P, Knolle PA, Unger K, Zender L, Nieswandt B, Gawaz M, Weber A*, Heikenwalder M* (2019) Platelet GPIbα is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat Med 25:641-655.
• Ringelhan M, Pfister D, O'Connor T, Pikarsky E, Heikenwalder M (2018) The immunology of hepatocellular carcinoma. Nat Immunol 19:222-232.
• Boege Y, Malehmir M, Healy ME, Bettermann K, Lorentzen A, Vucur M, Ahuja AK, Böhm F, Mertens JC, Shimizu Y, Frick L, Remouchamps C, Mutreja K, Kähne T, Sundaravinayagam D, Wolf MJ, Rehrauer H, Koppe C, Speicher T, Padrissa-Altés S, Maire R, Schattenberg JM, Jeong JS, Liu L, Zwirner S, Boger R, Hüser N, Davis RJ, Müllhaupt B, Moch H, Schulze-Bergkamen H, Clavien PA, Werner S, Borsig L, Luther SA, Jost PJ, Weinlich R, Unger K, Behrens A, Hillert L, Dillon C, Di Virgilio M, Wallach D, Dejardin E, Zender L, Naumann M, Walczak H, Green DR, Lopes M, Lavrik I, Luedde T, Heikenwalder M*, Weber A* (2017) A dual role of caspase-8 in triggering and sensing proliferation-associated DNA damage, a key determinant of liver cancer development. Cancer Cell 32:342-359.