top of page

PUBLICATIONS

Journals 

Editor of special editions

  • Vinken M., Scarino M.L., Roggen E., Blaauboer B. (2014) “Proceedings of the 17th Conference of the European Society for Toxicology”. Special edition of Toxicology In Vitro 28 (1).

  • Vinken M., Blaauboer B. (2015) “Proceedings of the 18th Conference of the European Society for Toxicology In Vitro”. Special edition of Toxicology In Vitro 29 (6).

  • Vinken M. (2015) “Connexin and pannexin signaling in organ functionality”. Special edition of Cellular and Molecular Life Sciences 72 (15).

  • Vinken M. (2017) “Adverse outcome pathways as versatile tools in in vitro and in silico toxicology: part I”. Special edition of Applied In Vitro Toxicology 3 (3).

  • Vinken M. (2017) “Adverse outcome pathways as versatile tools in in vitro and in silico toxicology: part II”. Special edition of Applied In Vitro Toxicology 3 (4).

  • Vinken M., Suter-Dick L., Kandarova H. (2019) “Proceedings of the 20th ESTIV congress 2018”. Toxicology In Vitro 60.

  • Vinken M. (2019) "Mitochondrial toxicity". Special edition of Applied In Vitro Toxicology 5.

  • Vinken M. (2021) “Connexin and pannexin signaling in health and disease”. Special edition of International Journal of Molecular Sciences 21-22.        

  • Vinken M. (2022) “Connexin and pannexin signaling in health and disease 2.0”. Special edition of International Journal of Molecular Sciences 22-23.      

  • Vinken M. (2022) “Molecular mechanisms of specific target organ toxicity”. Special edition of International Journal of Molecular Sciences 23.

  • Vinken M. (2023) “Connexin and pannexin signaling in health and disease 3.0”. Special edition of International Journal of Molecular Sciences 24.      

  • Vinken M. (2023) “Molecular mechanisms of specific target organ toxicity 2.0”. Special edition of International Journal of Molecular Sciences 24.

  • Vinken M. (2024) “New approach methodologies: from conceptual development to regulatory application”. Toxicology 499-505.

First author

  • Vinken M., Vanhaecke T., Rogiers V. (2003) De rol van intercellulaire communicatie via gap junctions bij ziekte. Nederlands Tijdschrift voor Geneeskunde 147: 2463-2466.

  • Vinken M., Papeleu P., Rogiers V., Vanhaecke T. (2006) Histone deacetylase inhibitors as potent modulators of cellular contacts. Current Drug Targets 7: 773-787.

  • Vinken M., Vanhaecke T., Papeleu P., Snykers S., Henkens T., Rogiers V. (2006) Connexins and their channels in cell growth and cell death. Cellular Signalling 18: 592-600.

  • Vinken M., Papeleu P., Snykers S., De Rop E., Henkens T., Chipman J.K., Rogiers V., Vanhaecke T. (2006) Involvement of cell junctions in hepatocyte culture functionality. Critical Reviews in Toxicology 36: 299-318.

  • Vinken M., Henkens T., Vanhaecke T., Papeleu P., Geerts A., Van Rossen E., Chipman J.K., Meda P., Rogiers V. (2006) Trichostatin A enhances gap junctional intercellular communication in primary cultures of adult rat hepatocytes. Toxicological Sciences 91: 484-492.

  • Vinken M., Vanhaecke T., Papeleu P., Snykers S., Henkens T., Elaut G., Chipman J.K., Rogiers V. (2006) A study of connexin expression in monolayer cultures of primary rat hepatocytes. Alternatives to Animal Experimentation 23: 456-460.

  • Vinken M., Henkens T., Snykers S., Lukaszuk A., Tourwé D., Rogiers V., Vanhaecke T. (2007) The novel histone deacetylase inhibitor 4-Me2N-BAVAH differentially affects cell junctions between primary hepatocytes. Toxicology 236: 92-102.

  • Vinken M., Henkens T., De Rop E., Fraczek J., Vanhaecke T., Rogiers V. (2008) Biology and pathobiology of gap junctional channels in hepatocytes. Hepatology 43: 1077-1088.

  • Vinken M., Vanhaecke T., Rogiers V. (2008) Junctional structures and hepatocellular carcinoma: from the lab to the clinic? Liver International 28: 432-434.

  • Vinken M., Doktorova T., Ellinger-Ziegelbauer H., Ahr H.J., Lock E., Carmichael P., Roggen E., van Delft J., Kleinjans J., Castell J., Bort R., Donato T., Ryan M., Corvi R., Keun H., Ebbels T., Atersuch T., Sansone S.-A., Rocca-Serra P., Stierum R., Jennings P., Pfaller W., Gmuender H., Vanhaecke T., Rogiers V. (2008) The carcinoGENOMICS project: critical selection of model compounds for the development of omics-based in vitro carcinogenicity screening assays. Mutation Research Reviews 629: 202-210.

  • Henkens T.*, Vinken M.*, Lukaszuk A., Tourwé D., Vanhaecke T., Rogiers V. (2008) Differential effects of hydroxamate histone deacetylase inhibitors on cellular functionality and gap junctions in primary cultures of mitogen-stimulated hepatocytes. Toxicology Letters 178: 37-43 (*Equal contribution).

  • Vinken M., De Rop E., Decrock E., De Vuyst E., Leybaert L., Vanhaecke T., Rogiers V. (2009) Epigenetic regulation of gap junctional intercellular communication: more than a way to keep cells quiet? Biochimica et Biophysica Acta Reviews on Cancer 1795: 53-61.

  • Vinken M., Decrock E., De Vuyst E., Leybaert L., Vanhaecke T., Rogiers V. (2009) Biochemical characterization of an in vitro model of hepatocellular apoptotic cell death. Alternatives to Laboratory Animals 37: 209-218.

  • Vinken M., Doktorova T., Decrock E., Leybaert L., Vanhaecke T., Rogiers V. (2009) Gap junctional intercellular communication as a target for liver toxicity and carcinogenicity. Critical Reviews in Biochemistry and Molecular Biology 44: 201-222.

  • Decrock E.*, Vinken M.*, De Vuyst E., Krysko D.V., D'Herde K., Vanhaecke T., Vandenabeele P., Rogiers V., Leybaert L. (2009) Connexin-related signalling in cell death: to live or let die? Cell Death and Differentiation 16: 524-536 (*Equal contribution).

  • Vinken M., Decrock E., De Vuyst E., De Bock M., Vandenbroucke R.E., De Geest B.G., Demeester J., Sanders N.N., Vanhaecke T., Leybaert L., Rogiers V. (2010) Connexin32 hemichannels contribute to the apoptotic-to-necrotic transition during Fas-mediated hepatocyte cell death. Cellular and Molecular Life Sciences 67: 907-918.

  • Vinken M., Vanhaecke T., Rogiers V. (2010) Emerging roles of connexin hemichannels in gastrointestinal and liver pathophysiology. World Journal of Gastrointestinal Pathophysiology 1: 115-117.

  • Vinken M., Decrock E., Leybaert L., Vanhaecke T., Rogiers V. (2010) Proteolytic cleavage of adherens junction components during Fas-dependent cell death in primary cultures of rat hepatocytes. Alternatives to Animal Experimentation 27: 151-157.

  • Vinken M., Ceelen L., Vanhaecke T., Rogiers V. (2010) Inhibition of gap junctional intercellular communication by toxic metals. Chemical Research in Toxicology 23: 1862-1867.

  • Vinken M., Snykers S., Fraczek J., Decrock E., Leybaert L., Rogiers V., Vanhaecke T. (2010) DNA methyltransferase 3a expression decreases during apoptosis in primary cultures of hepatocytes. Toxicology in Vitro 24: 445-451.

  • Vinken M., Decrock E., De Vuyst E., Ponsaerts R., D’hondt C., Bultynck G., Ceelen L., Vanhaecke T., Leybaert L., Rogiers V. (2011) Connexins: sensors and regulators of cell cycling. Biochimica et Biophysica Acta Reviews on Cancer 1815: 13-25.

  • Vinken M. (2011) Mathieu Vinken’s work on the role of connexin-related signalling in hepatic homeostasis and its relevance for liver-based in vitro modelling. Pharmaceutica Analytica Acta 2: 101e.

  • Vinken M. (2011) Role of connexin-related signalling in hepatic homeostasis and its relevance for liver-based in vitro modelling. World Journal of Gastrointestinal Pathophysiology 2: 82-87.

  • Vinken M., Decrock E., Doktorova T., Ramboer E., De Vuyst E., Bultynck G., Vanhaecke T., Leybaert L., Rogiers V. (2011) Characterization of spontaneous cell death in monolayer cultures of primary hepatocytes. Archives of Toxicology 85: 1589-1596.

  • Vinken M., Decrock E., Bultynck G., Vanhaecke T., Leybaert L., Rogiers V. (2012) Connexin43 signalling contributes to spontaneous apoptosis in cultures of primary hepatocytes. Toxicological Sciences 125: 175-186.

  • Vinken M., Vanhaecke T., Rogiers V. (2012) Primary hepatocyte cultures as tools in drug development: quo vadis? Toxicology in Vitro 26: 541-544.

  • Vinken M., Pauwels M., Ates G., Vivier M., Vanhaecke T., Rogiers V. (2012) Screening of repeated dose toxicity data present in SCC(NF)P/SCCS safety evaluations of cosmetic ingredients. Archives of Toxicology 86: 405-412.

  • Vinken M., Decrock E., Leybaert L., Bultynck G., Himpens B., Vanhaecke T., Rogiers V. (2012) Non-channel functions of connexins in cell growth and cell death. Biochimica et Biophysica Acta Biomembranes 1818: 2002-2008.

  • Vinken M., Buyl K., Vanhaecke T., Rogiers V. (2012) Effects of RNA interference-mediated suppression of connexin43 production on the expression of differentiation markers in cultures of adult primary rat hepatocytes. Alternatives to Animal Experimentation 1: 155-164.

  • Vinken M. (2012) Gap junctions and non-neoplastic liver disease. Journal of Hepatology 57: 655-662.

  • Vinken M., De Kock J., Oliveira A.G., Menezes G.B., Cogliati B., Zaidan Dagli M.L., Vanhaecke T., Rogiers V. (2012) Modifications in connexin expression in liver development and cancer. Cell Communication and Adhesion 19: 55-62.

  • Vinken M. (2012) Connexins: mediators, targets and biomarkers of liver toxicity. Pharmaceutica Analytica Acta 3: 1000e121.

  • Vinken M., Maes M., Cavill R., Valkenborg D., Ellis J.K., Decrock E., Leybaert L., Staes A., Gevaert K., Oliveira A.G., Menezes G.B., Cogliati B., Zaidan Dagli M.L., Ebbels T.M.D., Witter E., Keun H.C., Vanhaecke T., Rogiers V. (2013) Proteomic and metabolomic responses to connexin43 silencing in primary hepatocyte cultures. Archives of Toxicology 87: 883-894.

  • Vinken M., Maes M., Vanhaecke T., Rogiers V. (2013) Drug-induced liver injury: mechanisms types and biomarkers. Current Medicinal Chemistry 20: 3011-3021.

  • Vinken M. (2013) The adverse outcome pathway concept: a pragmatic tool in toxicology. Toxicology 312: 158-165.

  • Vinken M., Landesmann B., Goumenou M., Vinken S., Shah I., Jaeschke H., Willett C., Whelan M., Rogiers V. (2013) Development of an adverse outcome pathway from drug-mediated bile salt export pump inhibition to cholestatic liver injury. Toxicological Sciences 136: 97-106.

  • Vinken M., Whelan M., Rogiers V. (2014) Adverse outcome pathways: hype or hope? Archives of Toxicology 88: 1-2.

  • Vinken M., Maes M., Oliveira A.G., Cogliati B., Marques P.E., Menezes G.B., Zaidan Dagli M.L., Vanhaecke T., Rogiers V. (2014) Primary hepatocytes and their cultures in liver apoptosis research. Archives of Toxicology 88: 199-212.

  • Vinken M., Scarino M.L., Roggen E., Blaauboer B.J. (2014) Proceedings of the 17th ESTIV congress 2012. Toxicology In Vitro 28: 1-2.

  • Vinken M. (2015) Connexin hemichannels: novel mediators of toxicity. Archives of Toxicology 89: 143-145.

  • Cogliati B.*, Vinken M.*, da Silva T.C., Araújo C.M.M., Aloia T.P.A., Chaible L.M., Mori C.M.C., Dagli M.L.Z. (2015) Connexin43 deficiency accelerates skin wound healing and extracellular matrix remodeling in mice. Journal of Dermatological Science 79: 50-56 (*Equal contribution).

  • Vinken M. (2015) Introduction: connexins, pannexins and their channels as gatekeepers of organ physiology. Cellular and Molecular Life Sciences 72: 2775-2778.

  • Vinken M. (2015) Adverse outcome pathways and drug-induced liver injury testing. Chemical Research in Toxicology 28: 1391-1397.

  • Vinken M. (2016) Regulation of connexin signaling by the epigenetic machinery. Biochimica et Biophysica Acta Gene Regulatory Mechanisms 1859: 262-268.

  • Vinken M., Blaauboer B. (2017) In vitro testing of basal cytotoxicity: establishment of an adverse outcome pathway from chemical insult to cell death. Toxicology In Vitro 39: 104-110.

  • Vinken M., Knapen D., Vergauwen L., Hengstler J.G., Angrish M., Whelan M. (2017) Adverse outcome pathways: a concise introduction for toxicologists. Archives of Toxicology 91: 3697-3707.

  • Vinken M. (2018) In vitro prediction of drug-induced cholestatic liver injury: a challenge for the toxicologist. Archives of Toxicology 92: 1909-1912.

  • Vinken M. (2018) Taking adverse outcome pathways to the next level. Toxicology In Vitro 50: A1-A2.

  • Vinken M., Hengstler J. (2018) Characterization of hepatocyte-based in vitro systems for reliable toxicity testing. Archives of Toxicology 92: 2981-2986.

  • Vinken M., Wallace H.M. (2019) Key performance indicators of the contemporary European academic toxicologist. Archives of Toxicology 93: 1775-1776.

  • Vinken M., Suter-Dick L., Kandarova H. (2019) ESTIV2018: 20th anniversary of the International Congress on In Vitro Toxicology. Toxicology In Vitro 60: 293-295.

  • Vinken M. (2019) Omics-based input and output in the development and use of adverse outcome pathways. Current Opinion in Toxicology 18: 8-12.

  • Vinken M., Kramer N., Allen T.E.H., Hoffmans Y., Thatcher N., Levorato S., Traussnig H., Schulte S., Boobis A., Thiel A., Rietjens I.M.C.M. (2020) The use of adverse outcome pathways in the safety evaluation of food additives. Archives of Toxicology 94: 959-966.

  • Vinken M. (2020) 3Rs toxicity testing and disease modelling projects in the European Horizon 2020 research and innovation program. EXCLI Journal 19: 775-784.

  • Vinken M. (2020) A putative AOP for pneumonia related to COVID-19. Archives of Toxicology 94: 3343-3345.

  • Vinken M. (2020) Liver adverse outcome pathways: what’s in for the hepatologist? Journal of Gastrointestinal and Liver diseases 29: 659-664.

  • Vinken M. (2021) Primary hepatocyte cultures for liver disease modeling. Current Opinion in Toxicology 25: 1-5.

  • Vinken M. (2021) COVID-19 and the liver: an adverse outcome pathway perspective. Toxicology 455: 152765.

  • Vinken M., Benfenati E., Busquet E., Castell J., Clevert D.-A., de Kok T., Dirven H., Fritsche E., Geris L., Gozalbes R., Hartung T., Jennen D., Jover R., Kandarova H., Kramer N., Krul C., Luechtefeld T., Masereeuw R., Roggen E., Schaller S., Vanhaecke T., Yang C., Piersma A.H. (2021) Safer chemicals using less animals: kick-off of the European ONTOX project. Toxicology 458: 152846.

  • Vinken M. (2022) Toxic talk: pannexin1 channel communication as an emerging mechanism of toxicity. Toxicology 478: 153295.

  • Vinken M. (2022) Hepatology in space: effects of spaceflight and simulated microgravity on the liver. Liver International 42: 2599-2606.

 

Co-author

  • Adler S., Basketter D., Creton S., Pelkonen O., van Benthem J., Zuang V., Andersen K.E., Angers-Loustau A., Aptula A., Bal-Price A., Benfenati E., Bernauer U., Bessems J., Bois F.Y., Boobis A., Brandon E., Bremer S., Broschard T., Casati S., Coecke S., Corvi R., Cronin M., Daston G., Dekant W., Felter S., Grignard E., Gundert-Remy U., Heinonen T., Kimber I., Kleinjans J., Komulainen H., Kreiling R., Kreysa J., Leite S.B., Loizou G., Maxwell G., Mazzatorta P., Munn S., Pfuhler S., Phrakonkham P., Piersma A., Poth A., Prieto P., Repetto G., Rogiers V., Schoeters G., Schwarz M., Serafimova R., Tähti H., Testai E., van Delft J., van Loveren H., Vinken M., Worth A., Zaldivar J.M. (2011) Alternative (non-animal) methods for cosmetics testing: current status and future prospects-2010. Archives of Toxicology 85: 367-485.

  • Barnes D.A, Firman J.W., Belfield S.J., Cronin M.T.D., Vinken M., Janssen M.J., Masereeuw R. (2024) Development of an adverse outcome pathway network for nephrotoxicity. Archives of Toxicology 98: 929-942.

  • Bauwens M., Wimana L., Keyaerts M., Peleman C., Lahoutte T., Kerseman K., Snykers S., Vinken M., Mertens J., Bossuyt A. (2010) Preliminary in vivo evaluation of [131I]-2-I-D-Phe as a potential radionuclide therapeutic agent in R1M-fluc rhabdomyosarcoma tumour bearing NuNu mice using bioluminescent imaging. Cancer Biotherapy and Radiopharmaceuticals 25: 225-231.

  • Bavli D., Prill S., Ezra E., Levy G., Vinken M., Vanfleteren J., Jaeger M.S., Nahmias Y. (2016) Real-time monitoring of metabolic function in liver-on-chip microdevices tracks the dynamics of mitochondrial dysfunction. Proceedings of the National Academy of Sciences USA 113: E2231-E2240.

  • Bolleyn J., Fraczek J., Vinken M., Lizarraga D., Gaj S., van Delft J., Rogiers V., Vanhaecke T. (2011) Effect of Trichostatin A on miR-122 expression in primary hepatocyte cultures. Toxicology in Vitro 25: 1173-1182.

  • Bolleyn J., De Kock J., Mertens H., Fraczek J., Vinken M., Govaere O., Roskams T., Rogiers V, Vanhaecke T. (2012) Liver-specific miR-122 does not affect mRNA levels of cytochrome P450 enzymes and nuclear receptors in primary rat hepatocyte cultures. Alternatives to Animal Experimentation 1: 165-172.

  • Bolleyn J., De Kock J., Rodrigues R.M., Vinken M., Rogiers V., Vanhaecke T. (2015) MicroRNAs as key regulators of xenobiotic biotransformation and drug response. Archives of Toxicology 89: 1523-1541.

  • Ceelen L., De Spiegelaere W., David M., De Craene J., Vinken M., Vanhaecke T., Rogiers V. (2011) Selection of reliable reference genes for gene expression study in the HepaRG cell line. Biochemical Pharmacology 81: 1255-1261.

  • Chaudhari U., Nemade H., Sureshkumar P., Vinken M., Ates G., Rogiers V., Hescheler J., Hengstler J.G., Sachinidis A. (2018) Functional cardiotoxicity assessment of cosmetic compounds using human-induced pluripotent stem cell-derived cardiomyocytes. Archives of Toxicology 92: 371-381.

  • Clerbaux L.-A.,  Amigó N., Amorim M.J., Bal-Price A., Batista Leite S., Beronius A., Bezemer G.F.G., Bostroem A.-C., Carusi A., Coecke S., Concha R., Daskalopoulos E.P., Debernardi F., Edrosa E., Edwards S.W., Filipovska J., Garcia-Reyero N., Gavins F.N.E., Halappanavar S., Hargreaves A.J., Hogberg H., Huynh M.T., Jacobson D., Josephs-Spaulding J., Kim Y.J., Kong H.J., Krebs C.E., Lam A., Landesmann B., Racanelli Layton A., Lee Y.O., Macmillan D.S., Mantovani A., Margiotta-Casaluci L., Martens M., Masereeuw R., Mayasich S.A., Mei L.M., Mortensen H., Munoz Pineiro A., Nymark P., Ohayon E., Ojasi J.M., Paini A., Parissis N., Parvatam S., Pistollato F., Sachana M., Birkelund Sørli J., Sullivan K.M., Sund J., Tanabe S., Tsaioun K., Vinken M., Viviani L., Waspe J., Willett C., Wittwehr C. (2022) COVID-19 through adverse outcome pathways: building networks to better understand the disease: report of the 3rd CIAO AOP design workshop. ALTEX 39: 322-335.

  • De Bock M., Culot M., Wang N., Bol M., Decrock E., De Vuyst E., da Costa A., Dauwe I., Vinken M., Simon A.M., Rogiers V., De Ley G., Evans W.H., Bultynck G., Dupont G., Cecchelli R., Leybaert L. (2011) Connexin channels provide a target to manipulate brain endothelial calcium dynamics and blood brain barrier permeability. Journal of Cerebral Blood Flow and Metabolism 31: 1942-1957.

  • De Bock M., Decrock E., Wang N., Bol M., Vinken M., Bultynck G., Leybaert L. (2014) The dual face of connexin-based astroglial Ca2+ communication: a key player in brain physiology and a prime target in pathology. Biochimica et Biophysica Acta Molecular Cell Research 1843: 2211-2232.

  • Debruyne C., Riggio J., Gustafson E., O’Sullivan D., Vinken M., Vanhaecke T., De Troyer O. (2020) Facilitating data curation: a solution developed in the toxicology domain. Proceedings of International Conference on Semantic Computing: 315-320.

  • Decrock E., De Vuyst E., Moorhem V., Van Laeken L., De Bock M., Vinken M., Rogiers V., D’Herde K., Evans W.H., Naus C.C., Leybaert L. (2009) Connexin43 hemichannels contribute to the propagation of apoptotic cell death in a rat C6 glioma cell model. Cell Death and Differentiation 16: 151-163.

  • Decrock E., Vinken M., Bol M., D’Herde K. Rogiers V., Vandenabeele P., Krysko D.V., Bultynck G., Leybaert L. (2011) Calcium and connexin-based intercellular communication, a deadly catch? Cell Calcium 50: 310-321.

  • Decrock E., Krysko D.V., Vinken M., Kaczmarek A., Crispino G., Bol M., Wang N., De Bock M., De Vuyst E., Christian C.C., Rogiers V., Vandenabeele P., Erneux C., Mammano F., Bultynck G., Leybaert L. (2012) Transfer of IP3 through gap junctions is critical, but not sufficient, for the spread of apoptosis. Cell Death and Differentiation 19: 947-957.

  • Decrock E., De Bock M., Wang N., Gadicherla A.K., Bol M., Delvaeye T., Vandenabeele P., Vinken M., Bultynck G., Krysko D.V., Leybaert L. (2013) IP3, a small molecule with a powerful message. Biochimica et Biophysica Acta Molecular Cell Research 1833: 1772-1786.

  • Decrock E., Hoorelbeke D., Ramadan R., Delvaeye T., De Bock M., Wang N., Krysko D.V., Bultynck G., Aerts A., Vinken M., Leybaert L. (2017) Calcium, oxidative stress and connexin channels, a harmonious orchestra dictating the response to radiotherapy treatment? Biochimicia et Biophysica Acta Molecular Cell Research 1864: 1099-1120.

  • Deferm N., De Vocht T., Qi B., Van Brantegem P., Gijbels E., Vinken M., de Witte P., Bouillon T., Annaert P. (2019) Current insights in the complexities underlying drug-induced cholestasis. Critical Reviews in Toxicology 49: 520-548.

  • De Muynck K., Vanderborght B., Gijbels E., De Ponti F.F., Van Welden S., Guilliams M., Scott C.L., Beschin A., Vinken M., Lefere S., Geerts A., Verhelst X., Van Vlierberghe H., Devisscher L. (2023) Kupffer cells contested as early drivers in the pathogenesis of primary sclerosing cholangitis. American Journal of Pathology 193: 366-379.

  • De Muynck K., Heyerick L., De Ponti F.F., Vanderborght B., Meese T., Van Campenhout S., Baudonck L., Gijbels E., Rodrigues P.M., Banales J.M., Vesterhuus M., Karlsen T.H., Scott C.L., Vinken M., Van der Linden M., Hoorens A., Van Dorpe J., Lefere S., Geerts A., Van Nieuwerburgh F., Verhelst X., Van Vlierberghe H., Devisscher L. (2024) Osteopontin expression characterizes bile duct associated macrophages and correlates with liver fibrosis severity in primary sclerosing cholangitis. Hepatology 79: 269-288.

  • De Vuyst E., Decrock E., De Bock M., Wang N., Vinken M., Van Moorhem M., Lai C., Culot M., Rogiers V., Cecchelli R., Naus C., Evans H., Leybaert L. (2009) Ca2+ regulation of connexin43 hemichannels in C6 glioma and glial cells. Cell Calcium 46: 176-187.

  • D'hondt C., Ponsaerts R., De Smedt H., Vinken M., De Vuyst E., De Bock M., Wang N., Rogiers V., Leybaert L., Himpens B., Bultynck G. (2011) Pannexin channels in ATP release and beyond: an unexpected rendez-vous at the endoplasmic reticulum. Cellular Signalling 23: 305-316.

  • D'hondt C., Iyyathurai J., Vinken M., Rogiers V., Leybaert L., Himpens B., Bultynck G. (2013) Regulation of connexin- and pannexin-based channels by posttranslational modifications. Biology of the Cell 105: 373-398.

  • Diemar M., Vinken M., Teunis M., Krul C., Busquet F., Zajac J., Kandarova H., Corvi R., Russo M., Kharina A., Bryndum L., Santillo M., Bloch D., Kucheryavenko O., Panagiotakos D., Rogiers V., Beekhuijzen M., Giusti A., Najjar A., Courage C., Koenig T., Kolle S., Boonen H., Dhalluin S., Boberg J., Mülle B. Kukic P., Ritskes-Hoitinga M., Grasselli E., Zietek T., Stoddart G., Heusinkveld H., Castell J., Benfenati E., Yang H., Perera S., Paini A., Kramer N., Hartung T., Janssen M., Fritsche E., Jennen D., Piumatti M., Rathman J. Marusczyk J., Milec L., Roggen E. (2024) Report of the first ONTOX Stakeholder Network meeting: digging under the surface of ONTOX together with the stakeholders. Alternatives to Laboratory Animals 52: 171-131.

  • Doktorova T., Pauwels M., Vinken M., Vanhaecke T., Rogiers V. (2012) Opportunities for an alternative integrating testing strategy for carcinogen hazard assessment. Critical Reviews in Toxicology 42: 91-106.

  • Doktorova T., Ellinger-Ziegerbauer, Vinken M., Vanhaecke T., van Delft J., Kleinjans J., Ahr H.-J., Rogiers V. (2012) Comparison of hepatocarcinogen-induced gene expression profiles in conventional primary rat hepatocytes with in vivo rat liver data. Archives of Toxicology 86: 1399-1411.

  • Doktorova T., Ellinger-Ziegerbauer, Vinken M., Vanhaecke T., van Delft J., Kleinjans J., Ahr H.-J., Rogiers V. (2012) Comparison of genotoxicant-modified transcriptomic responses in conventional and epigenetically-stabilized primary rat hepatocytes with in vivo rat liver data. Archives of Toxicology 86: 1703-1715.

  • Doktorova T.Y., Yildirimman R., Vinken M., Vilardell M., Vanhaecke T., Gmuender H., Bort R., Brolen G., Holmgren G., Li R., Chesne C., van Delft J., Kleinjans J., Castell J., Bjorquist P., Herwig R., Rogiers V. (2013) Transcriptomic responses generated by hepatocarcinogens in a battery of liver-based in vitro models. Carcinogenesis 34: 1393-1402.

  • Doktorova T.Y., Ates G., Vinken M., Vanhaecke T., Rogiers V. (2014) Way forward in case of a false positive in vitro genotoxicity result for a cosmetic substance? Toxicology In Vitro 28: 54-59.

  • Doktorova T.Y., Yildirimman R., Ceelen L., Vilardell M., Vanhaecke T., Vinken M., Ates G., Heymans A., Gmuender H., Bort R., Corvi R., Phrakonkham P., Li R., Mouchet N., Chesne C., van Delft J., Kleinjans J., Castell J., Herwig R., Rogiers V. (2014) Testing chemical carcinogenicity by using a transcriptomics HepaRG-based model? EXCLI Journal 13: 623-637.

  • Du K., Williams D., McGill M.R., Xie Y., Farhood A., Vinken M., Jaeschke H. (2013) The gap junction inhibitor 2-aminoethoxy-diphenyl-borate protects against acetaminophen hepatotoxicity by inhibiting P450 enzymes and c-jun N-terminal kinase activation. Toxicology and Applied Pharmacology 273: 484-491.

  • Elaut G., Török G., Vinken M., Laus G., Papeleu P., Tourwé D. Rogiers V. (2002) Major phase I biotransformation pathways of Trichostatin A in rat hepatocytes and in rat and human liver microsomes. Drug Metabolism and Disposition 30: 1320-1328.

  • Elaut G., Henkens T., Papeleu P., Snykers S., Vinken M., Vanhaecke T., Rogiers V. (2006) Molecular mechanisms underlying the dedifferentiation process of isolated hepatocytes and their cultures. Current Drug Metabolism 7: 629-660.

  • Ellis J.K., Chan P.H., Doktorova T., Athersuch T.J., Cavill R., Vanhaecke T., Rogiers V., Vinken M., Nicholson J.K., Ebbels T.M.D., Keun H.C. (2010) The effect of the histone deacetylase inhibitor Trichostatin A on the metabolome of cultured primary hepatocytes. Journal of Proteome Research 9: 413-419.

  • Eskes C., Sullivan K., Aardema M., Spielmann H., Hill E., Schoeters G., Curren R., Vinken M., Blaauboer B. (2012) Comparing the challenges in developing and implementing 3Rs alternative methods in Europe and United States: industrial and academic perspectives. Alternatives to Animal Experimentation 1: 205-209.

  • Firman J.W., Pestana C.B., Rathman J.F., Vinken M., Yang C., Cronin M.T.D. (2021) A robust, mechanistically based in silico structural profiler for hepatic cholestasis. Chemical Research In Toxicology 34: 641-655.

  • Fraczek J., van Grunsven L.A., Vinken M., Snykers S., Deleu S., Vanderkerken K., Vanhaecke T., Rogiers V. (2009) Histone deacetylase inhibition and the regulation of cell growth with particular reference to liver pathobiology. Journal of Cellular and Molecular Medicine 13: 2990-3005.

  • Fraczek J., Vinken M., Tourwé D., Vanhaecke T., Rogiers V. (2012) Synergetic effects of DNA demethylation and histone deacetylase inhibition in primary rat hepatocytes. Investigational New Drugs 30: 1715-1724.

  • Godoy P., Hewitt N.J., Albrecht U., Andersen M.E., Ansari N., Bhattacharya S., Bode J.G., Bolleyn J., Borner C., Böttger J., Braeuning A., Budinsky R.A., Burkhardt B., Cameron N.R., Camussi G., Cho C.S., Choi Y.J., Craig Rowlands J., Dahmen U., Damm G., Dirsch O., Donato M.T., Dong J., Dooley S., Drasdo D., Eakins R., Ferreira K.S., Fonsato V., Fraczek J., Gebhardt R., Gibson A., Glanemann M., Goldring C.E., Gómez-Lechón M.J., Groothuis G.M., Gustavsson L., Guyot C., Hallifax D., Hammad S., Hayward A., Häussinger D., Hellerbrand C., Hewitt P., Hoehme S., Holzhütter H.G., Houston J.B., Hrach J., Ito K., Jaeschke H., Keitel V., Kelm J.M., Kevin Park B., Kordes C., Kullak-Ublick G.A., Lecluyse E.L., Lu P., Luebke-Wheeler J., Lutz A., Maltman D.J., Matz-Soja M., McMullen P., Merfort I., Messner S., Meyer C., Mwinyi J., Naisbitt D.J., Nussler A.K., Olinga P., Pampaloni F., Pi J., Pluta L., Przyborski S.A., Ramachandran A., Rogiers V., Rowe C., Schelcher C., Schmich K., Schwarz M., Singh B., Stelzer E.H., Stieger B., Stöber R., Sugiyama Y., Tetta C., Thasler W.E., Vanhaecke T., Vinken M., Weiss T.S., Widera A., Woods C.G., Xu J.J., Yarborough K.M., Hengstler J.G. (2013) Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Archives of Toxicology 87: 1315-1530.

  • Gomes Espírito Santo S., Da Silva T.C., Vinken M., Cogliati B., Barbisan L.F., Ribeiro Romualdo G. (2022) The implications of connexin 43 deficiency during the early stages of chemically induced mouse colon carcinogenesis. Antioxidants 11: 2368.

  • Gray A., Vinken M., Blaauboer B. (2015) Making sense of in vitro methods: proceedings of the 18th ESTIV congress. Toxicology In Vitro 29: 1215-1216.

  • Grinberg M., Stöber R.M., Edlund K., Rempel E., Godoy P., Reif R., Widera A., Madjar K., Schmidt-Heck W., Marchan R., Sachinidis A., Spitkovsky D., Hescheler J., Carmo H., Arbo M.D., van de Water B., Wink S., Vinken M., Rogiers V., Escher S., Hardy B., Mitic D., Myatt G., Waldmann T., Mardinoglu A., Damm G., Seehofer D., Nüssler A., Weiss T.S., Oberemm A., Lampen A., Schaap M.M., Luijten M., van Steeg H., Thasler W.E., Kleinjans J.C., Stierum R.H., Leist M., Rahnenführer J., Hengstler J.G. (2014) Toxicogenomics directory of chemically exposed human hepatocytes. Archives of Toxicology 88: 2261-2287.

  • Henkens T., Papeleu P., Elaut G., Vinken M., Rogiers V., Vanhaecke T. (2007) Trichostatin A, a critical factor in maintaining the functional differentiation of primary cultured rat hepatocytes. Toxicology and Applied Pharmacology 218: 64-71.

  • Henkens T., Vinken M., Vanhaecke T., Rogiers V. (2007) Modulation of CYP1A1 and CYP2B1 expression upon cell cycle progression in cultures of primary rat hepatocytes. Toxicology in Vitro 21: 1253-1257.

  • Henkens T.*, Snykers S.*, Vinken M., Lukaszuk A., Tourwé D., Verheyen G., Van Gompel J., Vanparys P., Rogiers V., Vanhaecke T. (2011) Preservation of hepatocellular functionality in cultures of primary rat hepatocytes upon exposure to 4-Me2N-BAVAH, a hydroxamate-based HDAC-inhibitor. Toxicology in Vitro 25: 100-109 (*Equal contribution).

  • Heremans J., Chevillard L., Mannes M., Mangialetto J., Leroy K., White J.F., Lamouroux A., Vinken M., Gardinet J., Van Mele B., Van den Brande N., Hoogenboom R., Madder A., Caveliers V., Mégarbane B., Hernot S., Ballet S., Martin C. (2022) Impact of doubling peptide length on in vivo hydrogel stability and sustained drug release. Journal of Controlled Release 350: 514-524.

  • Hoffmann S., Aiassa E., Angrish M., Beauso leil C., Bois F.Y., Ciccolallo L., Craig P.S., de Vries R.M.B., Dorne J.L.C.M. , Druwe I.L., Edwards S.W., Eskes C., Georgiadis M., Hartung T., Kienzler A., Kristjansson E.A., Lam J., Martino L., Meek B., Morgan R.L., Munoz-Guajardo I., Noyes P.D., Parmelli E., Piersma A., Rooney A., Sena E., Sullivan K., Tarazona J., Terron A., Thayer K., Turner J., Verbeek J., Verloo D., Vinken M., Watford S., Whaley P., Wikoff D., Willett K., Tsaioun K. (2022) Application of evidence-based methods to construct mechanism-driven chemical assessment frameworks. ALTEX 39: 499-518.

  • Horvat T., Landesmann B., Lostia A., Vinken M., Munn S., Whelan M. (2017) Adverse outcome pathway development from protein alkylation to liver fibrosis. Archives of Toxicology 91: 1523-1543.

  • Lamouroux A., Tournier M., Iaculli D., Caufriez A., Rusiecka O., Martin C., Bes V., Carpio L.E., Girardin Y., Loris R., Tabernilla A., Molica F., Gozalbes R. Mayan M., Vinken M., Kwak B., Ballet S. (2023) Structure-based design and synthesis of stapled 10Panx1 analogs for use in cardiovascular inflammatory diseases. Journal of Medicinal Chemistry 66: 13086-13102.

  • Leist M., Ghallab A., Graepel R., Marchan R., Hassan R., Bennekou S.H., Limonciel A., Vinken M., Schildknecht S., Waldmann T., Danen E., van Ravenzwaay B., Kamp H., Gardner I., Godoy P., Bois F.Y., Braeuning A., Reif R., Oesch F., Drasdo D., Höhme S., Schwarz M., Hartung T., Braunbeck T., Beltman J., Vrieling H., Sanz F., Forsby A., Gadaleta D., Fisher C., Kelm J., Fluri D., Ecker G., Zdrazil B., Terron A., Jennings P., van der Burg B., Dooley S., Meijer A.H., Willighagen E., Martens M., Evelo C., Mombelli E., Taboureau O., Mantovani A., Hardy B., Koch B., Escher S., van Thriel C., Cadenas C., Kroese D., van de Water B., Hengstler J.G. (2017) Adverse outcome pathways: opportunities, limitations and open questions. Archives of Toxicology 91: 3477-3505.

  • Mahony C.,  Ashton R.S., Birk B., Boobis A.R., Cull T., Daston G.P., Ewart L., Knudsen T.B., Manou I., Maurer-Stroh S., Margiotta-Casaluci L., Müller B.P., Nordlund P., Roberts R.A., Steger-Hartmann T., Vandenbossche E., Viant M.R., Vinken M., Whelan M., Zvonimir Z., Cronin M.T.D. (2020) New ideas for non-animal approaches to predict repeated-dose systemic toxicity: report from an EPAA blue sky workshop. Regulatory Toxicology and Pharmacology 114: 104668.

  • Moreno-Torres M., López E., Rapisarda A., Quintás G., Drees A., Steffensen I.-L., Luechtefeld T., Serrano-Candelas E., Garcia de Lomana M., Gadaleta D., Dirven H., Vinken M., Jover R. (2024) Patient and drug classifications and outcome prediction in drug-induced cholestasis. Analysis of 432 cases from literature review with machine learning support. Biomedicine & Pharmacotherapy 174: 116530.

  • Nelms M.D., Ates G., Madden J.C., Vinken M., Cronin M.T., Rogiers V., Enoch S.J. (2015) Proposal of an in silico profiler for categorisation of repeat dose toxicity data of hair dyes. Archives of Toxicology 89: 733-741.

  • Oliveira A.G., Marques P.E., Amaral S.S., Quintao J.L., Cogliati B., Zaidan Dagli M.L., Rogiers V., Vanhaecke T., Vinken M., Menezes G.B. (2013) Purinergic signaling in sterile liver injury. Liver International 33: 353-361.

  • Oorts M., Van Brantegem P., Deferm N., Chatterjee S., Dreesen E., Cooreman A., Vinken M., Richert L., Annaert P. (2021) Bosentan alters endo- and exogenous bile salt disposition in sandwich-cultured human hepatocytes. Journal of Pharmacology and Experimental Therapeutics 379: 20-32.

  • Papeleu P., Vanhaecke T., Elaut G., Vinken M., Henkens T., Snykers S., Rogiers V. (2005) Differential effects of histone deacetylase inhibitors in tumor and normal cells: what is the toxicological relevance? Critical Reviews in Toxicology 35: 363-378.

  • Papeleu P., Wullaert A., Elaut G., Henkens T., Vinken M., Laus G., Tourwé D., Beyaert R., Rogiers V., Vanhaecke T. (2007) Inhibition of NF-κβ activation by the histone deacetylase inhibitor 4-Me2N-BAVAH induces an early G1 cell cycle arrest in primary hepatocytes. Cell Proliferation 40: 640-655.

  • Prata Bacil G., Ribeiro Romualdo G., Piagge P.M.F.D., Cardoso D.R., Vinken M., Cogliati B., Barbisan L.F. (2023) Unraveling hepatic metabolomic profiles and morphological outcomes in a hybrid model of NASH in different mouse strains. Antioxidants 12: 290.

  • Punt A., Bouwmeester H., Blaauboer B.J., Coecke S., Hakkert B., Hendriks D.F.G, Jennings P., Kramer N.I., Neuhoff S., Masereeuw R., Paini A., Peijnenburg A.A.C.M., Rooseboom R., Shuler M.L., Sorrell I., Spee B., Strikwold M., van der Meer A.D., van der Zande M., Vinken M., Yang H., Bos P.M.J., Heringa M.B. (2020) New approach methodologies (NAMs) for human-relevant biokinetics predictions: meeting the paradigm shift in toxicology towards an animal-free chemical risk assessment. Alternatives to Animal Experimentation 37: 607-622.

  • Rodrigues R.M., De Kock J., Branson S., Vinken M., Govaere O., Roskams T., Meganathan K., Chaudari U., Sachinidis A., De Boe V., Vanhaecke T., Rogiers V. (2014) Human skin-derived stem cells as a novel cell source for in vitro hepatotoxicity screening of pharmaceuticals. Stem Cells and Development 23: 44-55.

  • Romualdo G.R., Rocha A.B., Vinken M., Cogliati B., Moreno F.S., Chaves M.A.G., Barbisan L.F. (2019) Drinking for protection? Epidemiological and experimental evidence on the beneficial effects of coffee or major coffee compounds against gastrointestinal and liver carcinogenesis. Food Research International 123:567-589.

  • Romualdo G.R., Silva E.A., da Silva T.C., Aloia T.P.A., Nogueira M.S., de Castro I.A., Vinken M., Barbisan L.F., Cogliati B. (2020) Burdock (Arctium lappa L.) root attenuates preneoplastic lesion development in a diet- and thioacetamide-induced model of steatohepatitis-associated hepatocarcinogenesis. Environmental Toxicology 35: 518-527.

  • Romualdo G.R., Prata G.B., da Silva T.C., Evangelista A.F., Reis R.M., Vinken M., Moreno F.S., Cogliati B., Barbisan L.F. (2020) The combination of coffee compounds attenuates early fibrosis-associated hepatocarcinogenesis in mice: involvement of miRNA profile modulation. Journal of Nutritional Biochemistry 85: 108479.

  • Romualdo G.R., Da Silva T.C., de Albuquerque Landi M.F., Morais J.A., Barbisan L.F., Vinken M., Oliveira C.P., Cogliati B. (2021) Sorafenib reduces steatosis-induced fibrogenesis in a human 3D co-culture model of non-alcoholic fatty liver disease. Environmental Toxicology 36: 168-176.

  • Romualdo G.R., Bacil Prata G., Sprocatti dos Santos A.C., Cardoso Valente L., Rodrigues J., Aparecida Marchesan Rodrigues M., Vinken M., Cogliati B., Barbisan L.F. (2022) A western diet-induced mouse model of non-alcoholic fatty liver disease associated to metabolic outcomes: features of gut microbiome-liver-adipose tissue axis. Nutrition 103-104: 111836.

  • Romualdo G.R., Cardoso Valente L., Sprocatti Dos Santos A.C., Alves Grandini N., Correa Camacho C.R., Vinken M., Cogliati B., Hou D.-X., Barbisan L.F. (2023) Effects of glyphosate exposure on western diet-induced non-alcoholic fatty liver disease in mice. Environmental Toxicology and Pharmacology 104: 104286.

  • Rogiers V., Vinken M., Snykers S., Henkens T., Fraczek J., De Rop E., De Kock J., Doktorova T., Lukaszuk A., Tourwé D., Vanhaecke T. (2008) Histone deacetylase inhibitors: opening a new field of research in the development of alternative methods? Alternatives to Laboratory Animals 14: 613-617.

  • Rosseel Z., Cortoos P.-J., Jonckheer J., Cools W., Vinken M., Reynaert H., De Waele E. (2023) Parenteral nutrition, sepsis, acute heart failure and hepatotoxic drugs are related to liver test disturbances in critically ill patients. Nutrients 15: 2612.

  • Rusiecka O.M., Montgomery J., Morel S., Batista-Almeida D., Van Campenhout R., Vinken M., Girao H., Kwak B.R. (2020) Canonical and non-canonical roles of connexin43 in cardioprotection. Biomolecules 10: e1225.

  • Schmeisser S., Miccoli A., von Bergen M., Berggren E., Braeuning A., Busch W., Desaintes C., Gourmelon A., Grafström R., Harrill J., Hartung T., Herzler M., Kass G., Kleinstreuer N., Leist M., Luijten M., Marx-Stoelting P., Poetz O., van Ravenzwaay B., Roggeband R., Rogiers V., Roth A., Sanders P., Thomas R.S., Vinggaard A.M., Vinken M., van de Water B., Luch A., Tralau T. (2023) New approach methodologies in human regulatory toxicology: not if, but how and when! Environment International 178: 108082.

  • Snykers S., Vanhaecke T., Papeleu P., Henkens T., Vinken M., Elaut G., Van Riet I., Rogiers V. (2006) In vitro multipotency of human bone marrow (mesenchymal) stem cells Alternatives to Animal Experimentation 23: 400-405.

  • Snykers S., Vanhaecke T., De Becker A., Papeleu P., Vinken M., Van Riet I., Rogiers V. (2007) Chromatin remodelling agent Trichostatin A: a key-factor in the endogenic differentiation of human mesenchymal stem cells. BMC Developmental Biology 7: 24.

  • Snykers S., Vinken M., Rogiers V., Vanhaecke T. (2007) Differential role of epigenetic modulators in malignant and normal stem cells: a novel tool in preclinical in vitro toxicology and clinical therapy. Archives of Toxicology 81: 533-544.

  • Snykers S., Henkens T. De Rop E., Vinken M., Fraczek J., De Kock J., De Prins E., Geerts A., Rogiers V., Vanhaecke T. (2009) The role of epigenetics in liver-specific gene transcription, hepatocyte differentiation, and stem cell reprogrammation. Journal of Hepatology 51: 187-211.

  • Sonawane A., Vadloori B., Poosala S., Kandarova H., Kulkarni M., Olayanju A., Dey T., Saxena U., Smirnova L., Kanda Y., Reddy J., Dravida S., Biswas S., Vinken M., Gettayacamin M., Ahluwalia A., Mondini F., Bhattacharya S., Kulkarni P., Jacobsen K., Vangala S., Millas A. (2022) Advances in animal models and cutting-edge research in alternatives: proceedings of the second international conference on 3Rs research and progress, Hyderabad, 2021. ATLA 50: 156-171.

  • Stefanska B., Vinken M., Szyf M. (2012) Epigenetics in toxicology: the implications of epigenetic alterations driven by external exposures for human health. Alternatives to Animal Experimentation 1: 173-185.

  • Svendsen C., Whaley P., Vist G.E., Husøy T., Beronius A., Di Consiglio E., Druwe I., Hartung T., Hatzi V.I., Hoffmann S., Hooijmans C.R., Machera K., Robinson J.F., Roggen E., Rooney A.A., Roth N., Spilioti E., Spyropoulou A., Tcheremenskaia O., Testai E., Vinken M., Mathisen G.H. (2023) Protocol for designing INVITES-IN, a tool for assessing the internal validity of in vitro studies. Evidence-Based Toxicology 1: 1-15.

  • Van Campenhout R., Muyldermans S., Vinken M., Devoogdt N., De Groof T.W.M. (2021) Therapeutic nanobodies targeting cell plasma membrane transport proteins: a high-risk/high-gain endeavor. Biomolecules 11: 63.

  • Vandenbroucke R.E., De Geest B.G., Bonné S., Vinken M., Vanhaecke T., Heimberg H. Wagner E., Rogiers V., De Smedt S.C., Demeester J., Sanders N.N. (2008) Prolonged gene silencing in hepatoma cells and primary hepatocytes after siRNA delivery with biodegradable poly-β-amino esters. Journal of Gene Medicine 10: 783-794.

  • Vanderborght B., De Muynck K., Gijbels E., Lefere S., Scott C., Guilliams M., Beschin A., Vinken M., Verhelst X., Geerts A., Van Vlierberghe H., Devisscher L. (2023) Transient Kupffer cell depletion and subsequent replacement by infiltrating monocyte-derived cells does not alter the induction or progression of hepatocellular carcinoma. International Journal of Cancer 152: 2615-2628.

  • Vanhaecke T., Vinken M., Henkens T., Snykers S., Papeleu P., Rogiers V. (2006) Effects of Trichostatin A on apoptosis-regulating proteins during hepatocyte isolation. Alternatives to Animal Experimentation 23: 441-444.

  • Vanhaecke T., Pauwels M., Vinken M., Ceelen L., Rogiers V. (2011) Towards a more realistic integrated in vitro strategy for repeated dose toxicity testing of cosmetic and pharmaceutical compounds. Archives of Toxicology 85: 365-366.

  • Walrave L., Vinken M., Albertini G., De Bundel D., Leybaert L., Smolders I. (2016) Inhibition of connexin43 hemichannels impairs spatial short-term memory without affecting spatial working memory. Frontiers in Cellular Neuroscience 10: 288.

  • Walrave L., Pierre A., Albertini G., Aourz N., De Bundel D., Van Eeckhaut A., Vinken M., Giaume C., Leybaert L., Smolders I. (2018) Inhibition of astroglial connexin43 hemichannels with TAT-Gap19 exerts anticonvulsant effects in rodents. Glia 66: 1788-1804.

  • Walrave L., Vinken M., Leybaert L., Smolders I. (2020) Astrocytic connexin43 channels as candidate anti-seizure drug targets. Biomolecules 10: 1578.

  • Wang N., De Bock M., Decrock E., Bol M., Gadicherla A., Vinken M., Rogiers V., Bukauskas, F., Bultynck G., Leybaert L (2013) Paracrine signaling through plasma membrane hemichannels. Biochimica et Biophysica Acta Biomembranes 1828: 35-50.

  • Wang N., De Vuyst E., Ponsaerts R., Boengler K., Palacios-Prado N., Wauman J., Lai C., De Bock M., Decrock E., Bol M., Vinken M., Rogiers V., Tavernier J., Evans W.H., Naus C.C., Bukauskas F.F. Sipido K.R., Heusch G., Schulz R., Bultynck G., Leybeart L. (2013) Selective inhibition of Cx43 hemichannels by Gap19 and its impact on myocardial ischemia/reperfusion injury. Basic Research in Cardiology 108: 309.

  • Willett C., Caverly Rae J., Goyak K.O., Minsavage G., Westmoreland C., Andersen M., Avigan M., Duché D., Hartung T., Jaeschke H., Kleensang A., Landesmann B., Toole C., Rowan A., Schultz T., Seed J., Senior  J., Shah I., Subramanian K., Vinken M., Watkins P. (2014) Building shared experience to advance practical application of pathway-based toxicology: liver toxicity mode-of-action. Alternatives to Animal Experimentation 31: 500-519.

  • Zgvac T., Hu T.-T., Van de Plas B., Vinken M., Ceulemans A.-G., Hachimi-Idrissi S., Sarre S., Michotte Y., Arckens L. (2013) Proteomic analysis of global protein expression changes in the endothelin-1 rat model for cerebral ischemia: rescue effect of mild hypothermia. Neurochemistry International 63: 379-388.

 

Last author

  • Ahmadian E., Eftekhari A., Samiei M., Maleki Dizaj S., Vinken M. (2019) The role and therapeutic potential of connexins, pannexins and their channels in Parkinson’s disease. Cellular Signalling 58: 111-118.

  • Arnesdotter E., Spinu N., Firman J., Ebbrell D., Cronin M.T.D., Vanhaecke T., Vinken M. (2021) Derivation, characterisation and analysis of an adverse outcome pathway network for human hepatotoxicity. Toxicology 459: 152856.

  • Arnesdotter E., Rogiers V., Vanhaecke T., Vinken M. (2021) An overview of current practices for regulatory risk assessment with lessons learnt from cosmetics in the European Union. Critical Reviews in Toxicology 51: 395-471.

  • Benoit L., Jornod F., Zgheib E., Tomkiewicz C., Koual M., Coustillet T., Barouki R., Audouze K., Vinken M.*, Coumoul X.* (2022) Adverse outcome pathway from activation of the AhR to breast cancer-related death. Environment International 165: 107323 (*Equal seniorship).

  • Busquet F., Vinken M. (2019) The use of social media in scientific research and creative thinking. Toxicology In Vitro 59: 51-54.

  • Caufriez A., Böck D., Martin C., Ballet S., Vinken M. (2020) Peptide-based targeting of connexins and pannexins for therapeutic purposes. Expert Opinion on Drug Discovery 15: 1213-1222.

  • Caufriez A., Tabernilla A., Van Campenhout R., Cooreman A., Leroy K., Sanz Serrano J., Kadam P., dos Santos Rodrigues B., Lamouroux A., Ballet S., Vinken M. (2022) Effects of drugs formerly suggested for COVID-19 repurposing on pannexin1 channels. International Journal of Molecular Sciences 23: 5664.

  • Caufriez A., Lamouroux A., Martin C., Iaculli D., Ince Ergüç E., Gozalbes R., Mayan M.D., Kwak B.R., Tabernilla A., Vinken M.*, Ballet S.* (2023) Determination of structural features that underpin the pannexin1 channel inhibitory activity of the peptide 10Panx1. Bioorganic Chemistry 138: 106612  (*Equal seniorship).

  • Ceelen L., Haesebrouck F., Vanhaecke T., Rogiers V., Vinken M. (2011) Modulation of connexin signalling by bacterial pathogens and their toxins. Cellular and Molecular Life Sciences 68: 3047-3064.

  • Ceelen L., Lieveld M., Forsyth R., Vinken M. (2013) The HepaRG cell line: a valuable in vitro tool for hepatitis virus infection studies. Hepatology International 7: 394-399.

  • Clippinger A.J., Allen D., Jarabek A.M., Corvaro M., Gaça M., Gehen S., Hotchkiss J.A., Patlewicz G., Melbourne M., Hinderliter P., Yoon M., Huh D., Lowit A., Buckley B., Bartels M., BéruBé K., Wilsong D.M., Indans I., Vinken M. (2018) Alternative approaches for acute inhalation toxicity testing to address global regulatory and non-regulatory data requirements: an international workshop report. Toxicology In Vitro 48: 53-70.

  • Cogliati B., Mennecier G., Willebrords J., da Silva T.C., Maes M., Veloso Alves Pereira I., Crespo Yanguas S., Hernandez-Blazques F.J., Zaidan Dagli M.L., Vinken M. (2016) Connexins, pannexins and their channels in fibroproliferative diseases. Journal of Membrane Biology 249: 199-213.

  • Cogliati B., Crespo Yanguas S., da Silva T.C., Aloia T.P.A., Nogueira M.S., Real-Lima M.A., Chaible L.M., Sanches D.S., Willebrords J., Maes M., Pereira I.V.A., de Castro I.A., Vinken M.*, Dagli M.L.Z.* (2016) Connexin32 deficiency exacerbates carbon tetrachloride-induced hepatocellular injury and liver fibrosis in mice. Toxicology Mechanisms and Methods 26: 362-370 (*Equal seniorship).

  • Cooreman A., Van Campenhout R., Ballet S., Annaert P., Van Den Bossche B., Colle I., Cogliati B., Vinken M. (2019) Connexin and pannexin (hemi)channels: emerging targets in the treatment of liver disease. Hepatology 69: 1317-1323.

  • Cooreman A., Van Campenhout R., Crespo Yanguas S., Gijbels E., Leroy K., Pieters A., Tabernilla A., Van Brantegem P., Annaert P., Cogliati B., Vinken M. (2020) Cholestasis differentially affects liver connexins. International Journal of Molecular Sciences 21: 6534.

  • Cooreman A., Caufriez A., Tabernilla A., Van Campenhout R., Leroy K., Kadam P., Sanz Serrano J., dos Santos Rodrigues B., Annaert P., Vinken M. (2022) Effects of drugs formerly proposed for COVID-19 treatment on connexin43 hemichannels. International Journal of Molecular Sciences 23: 5018.

  • Crespo Yanguas S., Cogliati B., Willebrords J., Maes M., Colle I., Van den Bossche B., Oliveira C.P., Andraus W., Alves V.A., Leclercq I., Vinken M. (2016) Experimental models of liver fibrosis. Archives of Toxicology 90: 1025-1048.

  • Crespo Yanguas S., Willebrords J., Maes M., da silva T.C., Veloso Alves Pereira I., Cogliati B., Dagli M.L., Vinken M. (2016) Connexins and pannexins in liver damage. EXCLI Journal 15: 177-186.

  • Crespo Yanguas S., Willebrords J., Johnstone S.R., Maes M., Decrock E., De Bock M., Leybaert L., Cogliati B., Vinken M. (2017) Pannexin1 as mediator of inflammation and cell death. Biochimica et Biophysica Acta Molecular Cell Research 1864: 51-61.

  • Crespo Yanguas S., da Silva T.C., Veloso Alves Pereira I., Willebrords J., Maes M., Sayuri Nogueira M., Alves de Castro I., Leclercq I., Romualdo G.R., Barbisan L.F., Leybaert L., Cogliati B.*, Vinken M.* (2018) TAT-Gap19 and carbenoxolone alleviate liver fibrosis in mice. International Journal of Molecular Sciences 19: 817 (*Equal seniorship).

  • Crespo Yanguas S., da Silva T.C., Veloso Alves Pereira I., Maes M., Willebrords J., Shestopalov V.I., Goes B.M., Sayuri Nogueira M., Alves de Castro I., Romualdo G.R., Barbisan L.F., Gijbels E., Vinken M.*, Cogliati B.* (2018) Genetic ablation of pannexin1 counteracts liver fibrosis in a chemical, but not in a surgical mouse model. Archives of Toxicology 92: 2607-2627 (*Equal seniorship).

  • Daston G.P., Mahony C., Thomas R.S., Vinken M. (2022) Assessing safety without animal testing: the road ahead. Toxicological Sciences 187: 214-218.

  • De Boever S., Devisscher L., Vinken M. (2024) Unraveling the micro- and nanoplastic predicament: a human-centric insight. Science of the Total Environment 916: 170262.

  • Desprez B., Birk B., Blaauboer, B., Boobis A., Carmichael P., Cronin M.T.D., Curie R., Daston G., Hubesch B., Jennings P., Klaric M., Kroese D., Mahony C., Ouédraogo G., Piersma A., Richarz. A.-N., Schwarz M., van Benthem J., van de Water B., Vinken M. (2019) A mode-of-action ontology model for safety evaluation of chemicals: outcome of a series of workshops on repeated dose toxicity. Toxicology In Vitro 59: 44-50.

  • dos Santos Rodrigues B., Leroy K., Mihajlovic M., De Boever S., Vanbellingen S., Cogliati B., Aerts J.L., Vinken M. (2023) Evaluation of functional candidate biomarkers of non-genotoxic hepatocarcinogenicity in human liver spheroid co-cultures. Archives of Toxicology 97: 1739-1751.

  • Fernando R.N., Chaudhari U., Escher S.E., Hengstler J.G. Hescheler J., Jennings P., Keun H.C., Kleinjans J.C.S., Kolde R., Kollipara L., Kopp-Schneider A., Limonciel A., Nemade H., Nguemo F., Peterson H., Prieto P., Rodrigues R.M., Sachinidis A., Schäfer C., Sickmann A., Spitkovsky D., Stöber R., van Breda S.G.J., van de Water B., Vivier M., Zahedi R.P., Vinken M.*, Rogiers V.* (2016) Watching the DETECTIVES: report of the general assembly of the EU project DETECTIVE, 24-25 November 2015. Archives of Toxicology 90: 1529-1539 (*Equal seniorship).

  • Fraczek J., Bolleyn J., Vanhaecke T., Rogiers V., Vinken M. (2013) Primary hepatocyte cultures for pharmaco-toxicological studies: at the busy crossroad of various anti-dedifferentiation strategies. Archives of Toxicology 87: 577-610.

  • Gijbels E., Vinken M. (2017) An update on adverse outcome pathways leading to liver injury. Applied In Vitro Toxicology 3: 283-285.

  • Gijbels E., Vilas-Boas V., Deferm N., Devisscher L., Jaeschke H., Annaert P., Vinken M. (2019) Mechanisms and in vitro models of drug-induced cholestasis. Archives of Toxicology 93: 1169-1186.

  • Gijbels E., Vilas-Boas V., Annaert P., Vanhaecke T., Devisscher L., Vinken M. (2020) Robustness testing and optimization of an adverse outcome pathway on cholestatic liver injury. Archives of Toxicology 94: 1151-1172 .

  • Gijbels E., Devisscher L., Vinken M. (2020) Dataset on transcriptomic profiling of cholestatic liver injury in an in vitro and in vivo animal model. Data in Brief 32: 106156.

  • Gijbels E., Pieters A., De Muynck K., Vinken M.*, Devisscher L.* (2021) Rodent models of cholestatic liver disease: a practical guide for translational research. Liver International 41: 656-682 (*Equal seniorship).

  • Gijbels E., Devisscher L., Vinken M. (2021) Testing in vitro tools for the prediction of cholestatic liver injury induced by non-pharmaceutical chemicals. Food and Chemical Toxicology 152: 112165.

  • Gijbels E., Devisscher L., Vinken M. (2021) Dataset on transcriptomic profiling of cholestatic liver injury induced by food additives and a cosmetic ingredient. Data in Brief 38: 107373.

  • Gijbels E., De Muynck K., Vanderborght B., Meese T., Van Nieuwerburgh F., Vanlander A., Berrevoet F., Hendrikx B., Hoorens A., Van Vlierberghe H., Vinken M.*, Devisscher L.* (2022) Systematic comparison of experimental and human obstructive cholestasis reveals conservation of canonical pathway activation and biomarkers relevant for cholestatic liver disease. Genes and Diseases 10: 18-21. *Equal seniorship.

  • Gustafson E., Debruyne C., De Troyer O., Rogiers V., Vinken M.*, Vanhaecke T.* (2020) Screening of repeated dose toxicity data in safety evaluation reports of cosmetic ingredients issued by the Scientific Committee on Consumer Safety between 2009 and 2019. Archives of Toxicology 94: 3723-3735. *Equal seniorship.

  • Jiang J., van Ertvelde J., Ertaylan G., Peeters R., Jennen D., de Kok T.M., Vinken M. (2023) Unraveling the mechanisms underlying drug-induced cholestatic liver injury: identifying key genes using machine learning techniques on human in vitro data sets. Archives of Toxicology 97: 2969-2981.

  • Kramer N.I., Hoffmans Y., Wu S., Thiel A., Thatcher N., Allen T.E.H., Levorato S., Traussnig H., Schulte S., Boobis A., Rietjens I.M.C.M., Vinken M. (2019) Characterizing the coverage of critical effects relevant in the safety evaluation of food additives by AOPs. Archives of Toxicology 93: 2115-2125.

  • Knudsen T.B., Vinken M. (2020) Excellence paves the way with current research in toxicology. Current Research in Toxicology 1: 1. 

  • Kunnen S.J., Arnesdotter E., Willenbockel C.T., Vinken M.*, van de Water B.* (2024) Qualitative and quantitative concentration-response modelling of gene co-expression networks to unlock hepatotoxic mechanisms for next generation chemical safety assessment. ALTEX 41: 213-232 (*Equal seniorship).

  • Leroy K., Pieters A., Tabernilla A., Cooreman A., Van Campenhout R., Cogliati B., Vinken M. (2020) Targeting gap junctional intercellular communication by hepatocarcinogenic compounds. Journal of Toxicology and Environmental Health, Part B: Critical Reviews 23: 255-275.

  • Leroy K., Pieters A., Cooreman A., Van Campenhout R. Cogliati B., Vinken M. (2021) Connexin-based channel activity is not specifically altered by hepatocarcinogenic chemicals. International Journal of Molecular Sciences 22: 11724.

  • Leroy K., Silva Costa C.J., Pieters A., dos Santos Rodrigues B., Van Campenhout R., Cooreman A., Tabernilla A., Cogliati B., Vinken M. (2021) Expression and functionality of connexin-based channels in human liver cancer cell lines. International Journal of Molecular Sciences 22: 12187.

  • Leroy K., Vilas-Boas V., Gijbels E., Vanderborght B., Devisscher L., Cogliati B., Van Den Bossche B., Colle I., Vinken M. (2022) Expression of connexins and pannexins in diseased human liver. EXCLI Journal 21: 1111-1129.

  • Limonciel A., Ates G., Carta G., Wilmes A., Watzele M., Shepard P.J., VanSteenhouse H.C., Seligmann B., Yeakley J.M., van de Water B, Vinken M.*, Jennings P.* (2018) Comparison of base-line and chemical-induced transcriptomic responses in HepaRG and RPTEC/TERT1 cells using TempO-Seq. Archives of Toxicology 92: 2517-2531 (*Equal seniorship).

  • Madden J.C., Rogiers V., Vinken M. (2014) Application of in silico and in vitro methods in the development of adverse outcome pathway constructs in wildlife. Philosophical Transactions of The Royal Society B Biological Sciences 369: 20130584.

  • Maes M., Decrock E., Cogliati B., Oliveira A.G., Marques P.E., Dagli M.L.Z., Menezes G.B., Mennecier G., Leybaert L., Vanhaecke T., Rogiers V., Vinken M. (2014) Connexin and pannexin (hemi)channels in the liver. Frontiers Physiology 4: 405.

  • Maes M., Crespo Yanguas S., Willebrords J., Cogliati B., Vinken M. (2015) Connexin and pannexin signaling in gastrointestinal and liver disease. Translational Research 166: 332-343.

  • Maes M., Cogliati B., Crespo Yanguas S., Willebrords J., Vinken M. (2015) Role of connexins and pannexins in digestive homeostasis. Cellular and Molecular Life Sciences 72: 2809-2821.

  • Maes M., Crespo Yanguas S., Willebrords J., Vinken M. (2015) Models and methods for in vitro testing of hepatic gap junctional communication. Toxicology In Vitro 30: 569-577.

  • Maes M., Vinken M.*, Jaeschke H.* (2016) Experimental models of hepatotoxicity  related to acute liver failure. Toxicology and Applied Pharmacology 290: 86-97 (*Equal seniorship).

  • Maes M., McGill M.R., da Silva T.C., Abels C., Lebofsky M., Monteiro de Araújo C.M., Tiburcio T., Veloso Alves Pereira I., Willebrords J., Crespo Yanguas S., Farhood A., Beschin A., Van Ginderachter J., Zaidan Dagli M.L., Jaeschke H., Cogliati B., Vinken M. (2016) Involvement of connexin43 in acetaminophen-induced liver injury. Biochimica et Biophysica Acta Molecular Basis of Disease 1862: 1111-1121.

  • Maes M., McGill M.R., da Silva T.C., Lebofsky M., Monteiro de Araújo C.M., Tiburcio T., Veloso Alves Pereira I., Willebrords J., Crespo Yanguas S., Farhood A., Zaidan Dagli M.L., Jaeschke H., Cogliati B., Vinken M. (2016) Connexin32: a mediator of acetaminophen-induced liver injury? Toxicology Mechanisms and Methods 26: 88-96.

  • Maes M., McGill M.R. , da Silva T.C, Abels C., Lebofsky M., Weemhoff J., Tiburcio T., Veloso Alves Pereira I., Willebrords J., Crespo Yanguas S., Farhood A., Beschin A., Van Ginderachter J.A., Penuela S., Jaeschke H., Cogliati B., Vinken M. (2017) Inhibition of pannexin1 channels alleviates acetaminophen-induced hepatotoxicity. Archives of Toxicology 91: 2245-2261.

  • Maes M., Vinken M. (2017) Connexin-based signaling and drug-induced hepatotoxicity. Journal of Clinical and Translational Research 3: 189-198.

  • Maes M., Crespo Yanguas S., Willebrords J., Weemhoff J.L., da Silva T.C., Decrock E., Lebofsky M., Pereira I.V.A., Leybaert L., Farhood A., Jaeschke J., Cogliati B., Vinken M. (2017) Connexin hemichannel inhibition reduces acetaminophen-induced liver injury in mice. Toxicology Letters 278: 30-37.

  • Mihajlovic M., Vinken M. (2022) Mitochondria as target of hepatotoxicity and drug-induced liver injury: molecular mechanisms and detection methods. International Journal of Molecular Sciences 23: 3315.

  • Mihajlovic M., Rosseel Z., de Waele E., Vinken M. (2024) Parenteral nutrition-associated liver injury: clinical relevance and mechanistic insights. Toxicological Sciences 199: 1-11.

  • Oredsson S., Coecke S., van der Valk J., Vinken M. (2019) What is understood by “animal-free research”? Toxicology In Vitro 57: 143-144.

  • Pieters A., Gijbels E., Cogliati B., Annaert P., Devisscher L., Vinken M. (2021) Biomarkers of cholestasis. Biomarkers in Medicine 15: 437-454.

  • Ramboer E., Vanhaecke T., Rogiers V., Vinken M. (2013) Primary hepatocyte cultures as prominent in vitro tools to study hepatic drug transporters. Drug Metabolism Reviews 45: 196-217.

  • Ramboer E., De Craene B., De Kock J., Vanhaecke T., Berx G., Rogiers V., Vinken M. (2014) Strategies for immortalization of primary hepatocytes. Journal of Hepatology 61: 925-943.

  • Ramboer E., Vanhaecke T., Rogiers V., Vinken M. (2015) Effects of Trichostatin A on drug uptake transporters in primary rat hepatocyte cultures. EXCLI Journal 14: 567-576.

  • Ramboer E., De Craene B., De Kock J., Berx G., Rogiers V., Vanhaecke T., Vinken M. (2015) Development and characterization of a new human hepatic cell line. EXCLI Journal 14: 875-889.

  • Rodrigues R.M. , Kollipara L., Chaudhari U., Sachinidis A., Zahedi R.P., Sickmann A., Kopp-Schneider A., Jiang X., Keun H., Hengstler J., Oorts M., Annaert P., Hoeben E., Gijbels E., De Kock J., Vanhaecke T., Rogiers V., Vinken M. (2018) Omics-based responses induced by bosentan in human hepatoma HepaRG cell cultures. Archives of Toxicology 92: 1939-1952.

  • Romualdo G.R., Leroy K., Silva Costa C.J., Bacil Prata G., Vanderborght B., da Silva T.C., Barbisan L.F., Andraus W., Devisscher L., Olsen Saraiva Câmara N., Vinken M.*, Cogliati B.* (2021) In vivo and in vitro models of hepatocellular carcinoma: current strategies for translational modeling. Cancers 13: 5583 (*Equal seniorship).

  • Samiei M., Ahmadian E., Eftekhari A., Ali Eghbal M., Rezaie F., Vinken M. (2019) Cell junctions and oral health. EXCLI Journal 18: 317-330.

  • Sanz-Serrano J., Callewaert E., De Boever S., Drees A., Verhoeven A., Vinken M. (2024) Chemical-induced liver cancer: an adverse outcome pathway perspective. Expert Opinion on Drug Safety 23: 425-438.

  • Tabernilla A., dos Santos Rodrigues B., Pieters A., Caufriez A., Leroy K., Van Campenhout R., Cooreman A., Gomes A.R., Arnesdotter E., Gijbels E., Vinken M. (2021) In vitro liver toxicity testing of chemicals: a pragmatic approach. International Journal of Molecular Sciences 22: 5038.

  • Tiburcio T.C, Willebrords J., da Silva T.C., Alves Pereira I.V., Nogueira M.S., Crespo Yanguas S., Maes M., dos Anjors Silva E., Dagli M.L., de Castro I.A., Oliveira C.P., Vinken M.*, Cogliati B.* (2017) Connexin32 deficiency is associated with liver injury, inflammation and oxidative stress in experimental non-alcoholic steatohepatitis. Clinical and Experimental Pharmacology and Physiology 44: 197-206 (*Equal seniorship).

  • Van Campenhout R., Crespo Yanguas S., Cooreman A., Gijbels E., Leroy K., Vilas-Boas V., Devoogdt N., Muyldermans S., Cogliati B, Vinken M. (2019) Increased expression of adherens junction components in mouse liver following bile duct ligation. Biomolecules 9: 636.

  • Van Campenhout R., Cooreman A., Leroy K., Rusiecka O.M., Van Brantegem P., Annaert P., Muyldermans S., Devoogdt N., Cogliati B., Kwak B.R., Vinken M. (2020) Non-canonical roles of connexins. Progress in Biophysics and Molecular Biology 153: 35-41.

  • Van Campenhout R., Gomes A.R., De Groof T.W.M., Muyldermans S., Devoogdt N., Vinken M. (2021) Mechanisms underlying connexin hemichannel activation in disease. International Journal of Molecular Sciences 22: 3503.

  • Van Campenhout R., Leroy K., Cooreman A., Tabernilla A., Cogliati B., Kadam P., Vinken M. (2022) Connexin-based channels in the liver. Comprehensive Physiology 12: 1-17.

  • Van Campenhout R., Cogliati B., Vinken M. (2023) Effects of acute and chronic disease on cell junctions in mouse liver. EXCLI Journal 22: 1-11.

  • Van Campenhout R., Caufriez A., Tabernilla A., Maerten A., De Boever S., Sanz Serrano J., Kadam P., Vinken M. (2023) Pannexin1 channels in the liver: an open enemy. Frontiers in Cell and Developmental Biology 11: 1220405.

  • Van Campenhout R., De Groof T.W.M., Kadam P., Kwak B.R., Muyldermans S., Devoogdt N., Vinken M. (2023) Nanobody-based pannexin1 channel inhibitors reduce inflammation in acute liver injury. Journal of Nanobiotechnology 21: 371.

  • van Ertvelde J., Verhoeven A., Maerten A., Cooreman A., dos Santos Rodrigues B., Sanz Serrano J., Mihajlovic M., Tripodi I., Teunis M., Jover R., Luechtefeld T., Vanhaecke T., Jiang J. Vinken M. (2023) Optimization of an adverse outcome pathway network on chemical-induced cholestasis using an artificial intelligence-assisted data collection and confidence level quantification. Journal of Biomedical Informatics 145: 104465.

  • Vilas-Boas V., Gijbels E., Cooreman A.,  Van Campenhout R., Gustafson E., Leroy K., Vinken M. (2019) Industrial, biocide and cosmetic chemical inducers of cholestasis. Chemical Research in Toxicology 32: 1327-1334.

  • Vilas-Boas V., Gijbels E., Jonckheer J., De Waele E., Vinken M. (2020) Cholestatic liver injury induced by food additives, dietary supplements and parenteral nutrition. Environment International 136: 105422.

  • Vilas-Boas V., Vinken M. (2021) Hepatotoxicity induced by nanomaterials: mechanisms and in vitro models. Archives of Toxicology 95: 27-52.

  • Vilas-Boas V., Gijbels E., Leroy K., Pieters A., Baze A., Parmentier C., Vinken M. (2021) Primary human hepatocyte spheroids as tools to study the hepatotoxic potential of non-pharmaceutical chemicals. International Journal of Molecular Sciences 22: 11005.

  • Willebrords J., Veloso Alves Pereira I., Maes M., Crespo Yanguas S., Colle I., Van Den Bossche B., Da silva T.C., Oliveira C.P., Andraus W., Alves V.A., Cogliati B., Vinken M. (2015) Strategies, models and biomarkers in experimental non-alcoholic fatty liver disease research. Progress in Lipid Research 59: 106-125.

  • Willebrords J., Crespo Yanguas S., Maes M., Decrock E., Wang E., Leybaert L., da Silva T.C., Veloso Alves Pereira I., Jaeschke H., Cogliati B, Vinken M. (2015) Structure, regulation and function of gap junctions in liver. Cell Communication and Adhesion 22: 29-37.

  • Willebrords J., Crespo Yanguas S., Maes M., Decrock E., Wang N., Leybaert L., Kwak B.R., Green C.R., Cogliati B., Vinken M. (2016) Connexins and their channels in inflammation. Critical Reviews in Biochemistry and Molecular Biology 51: 413-439.

  • Willebrords J., Cogliati B., Pereira I.V.A., da Silva T.C., Crespo Yanguas S., Maes M., Govoni V.M., Lima A., Felisbino D.A., Decrock E., Nogueira M.S., de Castro I.A., Leclercq I., Leybaert L., Rodrigues R.M., Vinken M. (2017) Inhibition of connexin hemichannels alleviates non-alcoholic steatohepatitis in mice. Scientific Reports 7: 8268.

  • Willebrords J., Maes M. Crespo Yanguas S., Vinken M. (2017) Inhibitors of connexin and pannexin channels as potential therapeutics. Pharmacology and Therapeutics 180: 144-160.

  • Willebrords J., Maes M., Veloso Alves Pereira I., da Silva T.C., Mollica Govoni V., Veras Lopes V., Crespo Yanguas S., Shestopalov V.I., Sayuri Nogueira M., Alves de Castro I., Farhood A., Mannaerts I., van Grunsven L., Akakpo J., Lebofsky M., Jaeschke H., Cogliati B., Vinken M. (2018) Protective effect of genetic deletion of pannexin1 in experimental mouse models of acute and chronic liver disease. Biochimica et Biophysica Acta Molecular Basis of Disease 1864: 819-830.

bottom of page