Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tamara Vanhaecke is active.

Publication


Featured researches published by Tamara Vanhaecke.


Stem Cells | 2009

In Vitro Differentiation of Embryonic and Adult Stem Cells into Hepatocytes: State of the Art

Sarah Snykers; Joery De Kock; Vera Rogiers; Tamara Vanhaecke

Stem cells are a unique source of self‐renewing cells within the human body. Before the end of the last millennium, adult stem cells, in contrast to their embryonic counterparts, were considered to be lineage‐restricted cells or incapable of crossing lineage boundaries. However, the unique breakthrough of muscle and liver regeneration by adult bone marrow stem cells at the end of the 1990s ended this long‐standing paradigm. Since then, the number of articles reporting the existence of multipotent stem cells in skin, neuronal tissue, adipose tissue, and bone marrow has escalated, giving rise, both in vivo and in vitro, to cell types other than their tissue of origin. The phenomenon of fate reprogrammation and phenotypic diversification remains, though, an enigmatic and rare process. Understanding how to control both proliferation and differentiation of stem cells and their progeny is a challenge in many fields, going from preclinical drug discovery and development to clinical therapy. In this review, we focus on current strategies to differentiate embryonic, mesenchymal(‐like), and liver stem/progenitor cells into hepatocytes in vitro. Special attention is paid to intracellular and extracellular signaling, genetic modification, and cell‐cell and cell‐matrix interactions. In addition, some recommendations are proposed to standardize, optimize, and enrich the in vitro production of hepatocyte‐like cells out of stem/progenitor cells. STEM CELLS 2009;27:577–605


Cell Death & Differentiation | 2009

Connexin-related signaling in cell death: to live or let die?

Elke Decrock; Mathieu Vinken; E De Vuyst; Dmitri V. Krysko; Katharina D'Herde; Tamara Vanhaecke; Peter Vandenabeele; Vera Rogiers; Luc Leybaert

Evidence is accumulating that some forms of cell death, like apoptosis, are not only governed by the complex interplay between extracellular and intracellular signals but are also strongly influenced by intercellular communicative networks. The latter is provided by arrays of channels consisting of connexin proteins, with gap junctions directly connecting the cytoplasm of neighboring cells and hemichannels positioned as pores that link the cytoplasm to the extracellular environment. The role of gap junctions in cell death communication has received considerable interest and recently hemichannels have joined in as potentially toxic pores adding their part to the cell death process. However, despite a large body of existing evidence, especially for gap junctions, the exact contribution of the connexin channel family still remains controversial, as both gap junctions and hemichannels may furnish cell death as well as cell survival signals. An additional layer of complexity is formed by the fact that connexin proteins as such, beyond their channel function, may influence the cell death process. We here review the current knowledge on connexins and their channels in cell death and specifically address the molecular mechanisms that underlie connexin-related signaling. We also briefly focus on pannexins, a novel set of connexin-like proteins that have been implicated in cellular responses to pathological insults.


Current Medicinal Chemistry | 2004

Trichostatin A-like hydroxamate histone deacetylase inhibitors as therapeutic agents: toxicological point of view.

Tamara Vanhaecke; Peggy Papeleu; Greetje Elaut; Vera Rogiers

Modulation of chromatin structure through histone acetylation/deacetylation is known to be one of the major mechanisms involved in the regulation of gene expression. Two opposing enzyme activities determine the acetylation state of histones: histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively acetylating or deacetylating the epsilon-amino groups of lysine residues located in the amino-terminal tails of the histones. In general, transcriptionally active chromatin is associated with hyperacetylated histones, whilst silenced chromatin is linked to hypoacetylated histones. A number of structurally divergent classes of HDAC inhibitors have been identified. They have been shown to induce cell cycle arrest, terminal differentiation and/or apoptosis in various cancer cell lines and inhibit tumor growth in animals. In particular, the reversible HDAC inhibitor Trichostatin A (TSA) and its hydroxamate analogues can effectively and selectively induce tumor growth arrest at very low concentrations (nano- to micromolar range). They form a group of so-called promising antitumor agents of which some are currently under clinical trial. Since the selection of a molecule for further drug development requires a balance of biological potency, safety and pharmacokinetics, it is of paramount importance to elucidate the pharmacokinetic and toxicological properties of these HDAC inhibitors before they can be considered as potential new drugs. Primary hepatocytes and their cultures are well-differentiated in vitro models and can be used to study simultaneously the biological effects of HDAC inhibitors and their biotransformation. The present review provides a state-of-the-art of our current knowledge of the pharmacological and toxicological effects on proliferating cells of TSA and its hydroxamate-based structural analogues. Besides a theoretical basis, an overview of the experimental results, obtained by the authors using primary rat hepatocytes as an in vitro model, is given.


Methods of Molecular Biology | 2006

Isolation of Rat Hepatocytes

Peggy Papeleu; Tamara Vanhaecke; Tom Henkens; Greetje Elaut; Mathieu Vinken; Sarah Snykers; Vera Rogiers

In vitro models, based on liver cells or tissues, are indispensable in the early preclinical phase of drug development. An important breakthrough in establishing cell models has been the successful high-yield preparation of intact hepatocytes. In this chapter, the practical aspects of the two-step collagenase perfusion method, modified from the original procedure of Seglen, are outlined. Although applicable to the liver of various species, including human, the practical aspects of the method are explained here for rat liver. Critical parameters for the successful isolation of primary rat hepatocytes are highlighted and a troubleshooting guide is provided. In addition, a new development based on the inhibition of histone deacetylase activity is presented. This approach allows inhibition of cell-cycle reentry during hepatocyte isolation, a process known to underlie the dedifferentiation process of cultured hepatocytes.


Hepatology | 2008

Biology and pathobiology of gap junctional channels in hepatocytes

Mathieu Vinken; Tom Henkens; Evelien De Rop; Joanna Fraczek; Tamara Vanhaecke; Vera Rogiers

The present review provides the state of the art of the current knowledge concerning gap junctional channels and their roles in liver functioning. In the first part, we summarize some relevant biochemical properties of hepatic gap junctional channels, including their structure and regulation. In the second part, we discuss the involvement of gap junctional channels in the occurrence of liver cell growth, liver cell differentiation, and liver cell death. We further exemplify their relevance in hepatic pathophysiology. Finally, a number of directions for future liver gap junctional channel research are proposed, and the up‐regulation of gap junctional channel activity as a novel strategy in (liver) cancer therapy is illustrated. (HEPATOLOGY 2007.)


Biochimica et Biophysica Acta | 2012

Non-channel functions of connexins in cell growth and cell death.

Mathieu Vinken; Elke Decrock; Luc Leybaert; Geert Bultynck; Bernard Himpens; Tamara Vanhaecke; Vera Rogiers

Cellular communication mediated by gap junction channels and hemichannels, both composed of connexin proteins, constitutes two acknowledged regulatory platforms in the accomplishment of tissue homeostasis. In recent years, an abundance of reports has been published indicating functions for connexin proteins in the control of the cellular life cycle that occur independently of their channel activities. This has yet been most exemplified in the context of cell growth and cell death, and is therefore as such addressed in the current paper. Specific attention is hereby paid to the molecular mechanisms that underpin the cellular non-channel roles of connexin proteins, namely the alteration of the expression of tissue homeostasis determinants and the physical interaction with cell growth and cell death regulators. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


BMC Developmental Biology | 2007

Chromatin remodeling agent trichostatin A: a key-factor in the hepatic differentiation of human mesenchymal stem cells derived of adult bone marrow

Sarah Snykers; Tamara Vanhaecke; Ann De Becker; Peggy Papeleu; Mathieu Vinken; Ivan Van Riet; Vera Rogiers

BackgroundThe capability of human mesenchymal stem cells (hMSC) derived of adult bone marrow to undergo in vitro hepatic differentiation was investigated.ResultsExposure of hMSC to a cocktail of hepatogenic factors [(fibroblast growth factor-4 (FGF-4), hepatocyte growth factor (HGF), insulin-transferrin-sodium-selenite (ITS) and dexamethasone)] failed to induce hepatic differentiation. Sequential exposure to these factors (FGF-4, followed by HGF, followed by HGF+ITS+dexamethasone), however, resembling the order of secretion during liver embryogenesis, induced both glycogen-storage and cytokeratin (CK)18 expression. Additional exposure of the cells to trichostatin A (TSA) considerably improved endodermal differentiation, as evidenced by acquisition of an epithelial morphology, chronological expression of hepatic proteins, including hepatocyte-nuclear factor (HNF)-3β, alpha-fetoprotein (AFP), CK18, albumin (ALB), HNF1α, multidrug resistance-associated protein (MRP)2 and CCAAT-enhancer binding protein (C/EBP)α, and functional maturation, i.e. upregulated ALB secretion, urea production and inducible cytochrome P450 (CYP)-dependent activity.ConclusionhMSC are able to undergo mesenchymal-to-epithelial transition. TSA is hereby essential to promote differentiation of hMSC towards functional hepatocyte-like cells.


Critical Reviews in Toxicology | 2006

Involvement of cell junctions in hepatocyte culture functionality.

Mathieu Vinken; Peggy Papeleu; Sarah Snykers; Evelien De Rop; Tom Henkens; James K. Chipman; Vera Rogiers; Tamara Vanhaecke

In liver, like in other multicellular systems, the establishment of cellular contacts is a prerequisite for normal functioning. In particular, well-defined cell junctions between hepatocytes, including adherens junctions, desmosomes, tight junctions, and gap junctions, are known to play key roles in the performance of liver-specific functionality. In a first part of this review article, we summarize the current knowledge concerning cell junctions and their roles in hepatic (patho)physiology. In a second part, we discuss their relevance in liver-based in vitro modeling, thereby highlighting the use of primary hepatocyte cultures as suitable in vitro models for preclinical pharmaco-toxicological testing. We further describe the actual strategies to regain and maintain cell junctions in these in vitro systems over the long-term.


Biochimica et Biophysica Acta | 2011

Connexins: sensors and regulators of cell cycling.

Mathieu Vinken; Elke Decrock; Elke De Vuyst; Raf Ponsaerts; Catheleyne D'hondt; Geert Bultynck; Liesbeth Ceelen; Tamara Vanhaecke; Luc Leybaert; Vera Rogiers

It is nowadays well established that gap junctions are critical gatekeepers of cell proliferation, by controlling the intercellular exchange of essential growth regulators. In recent years, however, it has become clear that the picture is not as simple as originally anticipated, as structural precursors of gap junctions can affect cell cycling by performing actions not related to gap junctional intercellular communication. Indeed, connexin hemichannels also foresee a pathway for cell growth communication, albeit between the intracellular compartment and the extracellular environment, while connexin proteins as such can directly or indirectly influence the production of cell cycle regulators independently of their channel activities. Furthermore, a novel set of connexin-like proteins, the pannexins, have lately joined in as regulators of the cell proliferation process, which they can affect as either single units or as channel entities. In the current paper, these multifaceted aspects of connexin-related signalling in cell cycling are reviewed.


Critical Reviews in Toxicology | 2005

Differential Effects of Histone Deacetylase Inhibitors in Tumor and Normal Cells—What Is the Toxicological Relevance?

Peggy Papeleu; Tamara Vanhaecke; Greetje Elaut; Mathieu Vinken; Tom Henkens; Sarah Snykers; Vera Rogiers

Histone deacetylase (HDAC) inhibitors target key steps of tumor development: They inhibit proliferation, induce differentiation and/or apoptosis, and exhibit potent antimetastatic and antiangiogenic properties in transformed cells in vitro and in vivo. Preliminary studies in animal models have revealed a relatively high tumor selectivity of HDAC inhibitors, strenghtening their promising potential in cancer chemotherapy. Until now, preclinical in vitro research has almost exclusively been performed in cancer cell lines and oncogene-transformed cells. However, as cell proliferation and apoptosis are essential for normal tissue and organ homeostasis, it is important to investigate how HDAC inhibitors influence the regulation of and interplay between proliferation, differentiation, and apoptosis in primary cells as well. This review highlights the discrepancies in molecular events triggered by trichostatin A, the reference compound of hydroxamic acid-containing HDAC inhibitors, in hepatoma cells and primary hepatocytes (which are key targets for drug-induced toxicity). The implications of these differential outcomes in both cell types are discussed with respect to both toxicology and drug development. In view of the future use of HDAC inhibitors as cytostatic drugs, it is highly recommended to include both tumor cells and their healthy counterparts in preclinical developmental studies. Screening the toxicological properties of compounds early in their development process, using a battery of different cell types, will enable researchers to discard those compounds bearing undesirable adverse activity before entering into expensive clinical trials. This will not only reduce the risk for harmful exposure of patients but also save time and money.

Collaboration


Dive into the Tamara Vanhaecke's collaboration.

Top Co-Authors

Avatar

Vera Rogiers

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Mathieu Vinken

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Pieter Jan Coenraads

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Maria Dusinska

Norwegian Institute for Air Research

View shared research outputs
Top Co-Authors

Avatar

Ulrike Bernauer

Federal Institute for Risk Assessment

View shared research outputs
Top Co-Authors

Avatar

Berit Granum

Norwegian Institute of Public Health

View shared research outputs
Top Co-Authors

Avatar

Janine Ezendam

Centre for Health Protection

View shared research outputs
Top Co-Authors

Avatar

Eric Gaffet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Joery De Kock

Vrije Universiteit Brussel

View shared research outputs
Researchain Logo
Decentralizing Knowledge