Network


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

Hotspot


Dive into the research topics where Daniel Benten is active.

Publication


Featured researches published by Daniel Benten.


Hepatology | 2011

Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells

Joerg Schrader; Timothy T. Gordon-Walker; Rebecca L. Aucott; Marielle Van Deemter; Alexander Quaas; Shaun V. Walsh; Daniel Benten; Stuart J. Forbes; Rebecca G. Wells; John P. Iredale

There is increasing evidence that the physical environment is a critical mediator of tumor behavior. Hepatocellular carcinoma (HCC) develops within an altered biomechanical environment, and increasing matrix stiffness is a strong predictor of HCC development. The aim of this study was to establish whether changes in matrix stiffness, which are characteristic of inflammation and fibrosis, regulate HCC cell proliferation and chemotherapeutic response. Using an in vitro system of “mechanically tunable” matrix‐coated polyacrylamide gels, matrix stiffness was modeled across a pathophysiologically relevant range, corresponding to values encountered in normal and fibrotic livers. Increasing matrix stiffness was found to promote HCC cell proliferation. The proliferative index (assessed by Ki67 staining) of Huh7 and HepG2 cells was 2.7‐fold and 12.2‐fold higher, respectively, when the cells were cultured on stiff (12 kPa) versus soft (1 kPa) supports. This was associated with stiffness‐dependent regulation of basal and hepatocyte growth factor–stimulated mitogenic signaling through extracellular signal‐regulated kinase, protein kinase B (PKB/Akt), and signal transducer and activator of transcription 3. β1‐Integrin and focal adhesion kinase were found to modulate stiffness‐dependent HCC cell proliferation. Following treatment with cisplatin, we observed reduced apoptosis in HCC cells cultured on stiff versus soft (physiological) supports. Interestingly, however, surviving cells from soft supports had significantly higher clonogenic capacity than surviving cells from a stiff microenvironment. This was associated with enhanced expression of cancer stem cell markers, including clusters of differentiation 44 (CD44), CD133, c‐kit, cysteine‐X‐cysteine receptor 4, octamer‐4 (CXCR4), and NANOG. Conclusion: Increasing matrix stiffness promotes proliferation and chemotherapeutic resistance, whereas a soft environment induces reversible cellular dormancy and stem cell characteristics in HCC. This has implications for both the treatment of primary HCC and the prevention of tumor outgrowth from disseminated tumor cells. (HEPATOLOGY 2011;)


Laboratory Investigation | 2005

ADAMTS13 Is expressed in hepatic stellate cells

Wenhua Zhou; Mari Inada; Tai Ping Lee; Daniel Benten; Sergey Lyubsky; Eric E. Bouhassira; Sanjeev Gupta; Han-Mou Tsai

ADAMTS13 is a circulating zinc metalloprotease that cleaves the hemostatic glycoprotein von Willebrand factor (VWF) in a shear-dependent manner. Deficiency in ADAMTS13, owing to genetic mutations or autoimmune inhibitors, causes thrombotic thrombocytopenic purpura (TPP). Northern blot analysis has shown that ADAMTS13 is expressed primarily in the liver. By using real-time RT-PCR, we confirmed that in mice the liver had the highest level of the ADAMTS13 transcript. To identify the liver cell-type-specific origin of ADAMTS13, we used in situ hybridization techniques to investigate the pattern of ADAMTS13 expression in the liver; analyzed the ADAMTS13 proteolytic activity in the culture media of fractionated liver cells; and confirmed ADAMTS13 expression with RT-PCR analysis and cloning of the mouse ADAMTS13 gene. The results revealed that ADAMTS13 was expressed primarily in cell fractions enriched in hepatic stellate cells. The mouse ADAMTS13 cloned from primary hepatic stellate cells was similar to its human counterpart in digesting VWF and was susceptible to suppression by EDTA or the IgG inhibitors of patients with TTP. Since hepatic stellate cells are believed to play a major role in the development of hepatic fibrosis and cirrhosis, the identification of the liver cell-type expressing ADAMTS13 will have important implications for understanding pathophysiological mechanisms regulating ADAMTS13 expression.


Blood | 2008

Simultaneous targeting of Aurora kinases and Bcr-Abl kinase by the small molecule inhibitor PHA-739358 is effective against imatinib-resistant BCR-ABL mutations including T315I

Artur Gontarewicz; Stefan Balabanov; Gunhild Keller; Riccardo Colombo; Alessio Graziano; Enrico Pesenti; Daniel Benten; Carsten Bokemeyer; Walter Fiedler; Jurgen Moll; Tim H. Brümmendorf

The emergence of resistance to imatinib (IM) mediated by mutations in the BCR-ABL domain has become a major challenge in the treatment of chronic myeloid leukemia (CML). Here, we report on studies performed with a novel small molecule inhibitor, PHA-739358, which selectively targets Bcr-Abl and Aurora kinases A to C. PHA-739358 exhibits strong antiproliferative and proapoptotic activity against a broad panel of human BCR-ABL-positive and -negative cell lines and against murine BaF3 cells ectopically expressing wild-type (wt) or IM-resistant BCR-ABL mutants, including T315I. Pharmacologic synergism of IM and PHA-739358 was observed in leukemia cell lines with subtotal resistance to IM. Treatment with PHA-739358 significantly decreased phosphorylation of histone H3, a marker of Aurora B activity and of CrkL, a downstream target of Bcr-Abl, suggesting that PHA-739358 acts via combined inhibition of Bcr-Abl and Aurora kinases. Moreover, strong antiproliferative effects of PHA-739358 were observed in CD34(+) cells derived from untreated CML patients and from IM-resistant individuals in chronic phase or blast crisis, including those harboring the T315I mutation. Thus, PHA-739358 represents a promising new strategy for treatment of IM-resistant BCR-ABL-positive leukemias, including those harboring the T315I mutation. Clinical trials investigating this compound in IM-resistant CML have recently been initiated.


Journal of Clinical Investigation | 2008

Transplanted endothelial cells repopulate the liver endothelium and correct the phenotype of hemophilia A mice

Antonia Follenzi; Daniel Benten; Phyllis M. Novikoff; Louisa Faulkner; Sanj Raut; Sanjeev Gupta

Transplantation of healthy cells to repair organ damage or replace deficient functions constitutes a major goal of cell therapy. However, the mechanisms by which transplanted cells engraft, proliferate, and function remain unknown. To investigate whether host liver sinusoidal endothelium could be replaced with transplanted liver sinusoidal endothelial cells, we developed an animal model of tissue replacement that utilized a genetic system to identify transplanted cells and induced host-cell perturbations to confer a proliferative advantage to transplanted cells. Under these experimental conditions, transplanted cells engrafted efficiently and proliferated to replace substantial portions of the liver endothelium. Tissue studies demonstrated that transplanted cells became integral to the liver structure and reacquired characteristic endothelial morphology. Characterization of transplanted endothelial cells by membrane markers and studies of cellular function, including synthesis and release of coagulation factor VIII, demonstrated that transplanted cells were functionally intact. Further analysis showed that repopulation of the livers of mice that model hemophilia A with healthy endothelial cells restored plasma factor VIII activity and corrected their bleeding phenotype. Our studies therefore suggest that transplantation of healthy endothelial cells should be considered for cell therapy of relevant disorders and that endothelial reconstitution with transplanted cells may offer an excellent paradigm for defining organ-specific pathophysiological mechanisms.


Nature Medicine | 2011

RGB marking facilitates multicolor clonal cell tracking

Kristoffer Weber; Michael Thomaschewski; Michael Warlich; T. Volz; Kerstin Cornils; Birte Niebuhr; Maike Täger; M. Lütgehetmann; J M. Pollok; Carol Stocking; M. Dandri; Daniel Benten; Boris Fehse

We simultaneously transduced cells with three lentiviral gene ontology (LeGO) vectors encoding red, green or blue fluorescent proteins. Individual cells were thereby marked by different combinations of inserted vectors, resulting in the generation of numerous mixed colors, a principle we named red-green-blue (RGB) marking. We show that lentiviral vector–mediated RGB marking remained stable after cell division, thus facilitating the analysis of clonal cell fates in vitro and in vivo. Particularly, we provide evidence that RGB marking allows assessment of clonality after regeneration of injured livers by transplanted primary hepatocytes. We also used RGB vectors to mark hematopoietic stem/progenitor cells that generated colored spleen colonies. Finally, based on limiting-dilution and serial transplantation assays with tumor cells, we found that clonal tumor cells retained their specific color-code over extensive periods of time. We conclude that RGB marking represents a useful tool for cell clonality studies in tissue regeneration and pathology.


Hepatology | 2016

Systemic inflammation in decompensated cirrhosis: Characterization and role in acute-on-chronic liver failure

Joan Clària; Rudolf E. Stauber; Minneke J. Coenraad; Richard Moreau; Rajiv Jalan; Marco Pavesi; Alex Amoros; Esther Titos; José Alcaraz-Quiles; Karl Oettl; Manuel Morales-Ruiz; Paolo Angeli; Marco Domenicali; Carlo Alessandria; Alexander L. Gerbes; Julia Wendon; Frederik Nevens; Jonel Trebicka; Wim Laleman; Faouzi Saliba; Tania M. Welzel; Agustín Albillos; Thierry Gustot; Daniel Benten; François Durand; Pere Ginès; Mauro Bernardi; Vicente Arroyo

Acute‐on‐chronic liver failure (ACLF) in cirrhosis is characterized by acute decompensation (AD), organ failure(s), and high short‐term mortality. Recently, we have proposed (systemic inflammation [SI] hypothesis) that ACLF is the expression of an acute exacerbation of the SI already present in decompensated cirrhosis. This study was aimed at testing this hypothesis and included 522 patients with decompensated cirrhosis (237 with ACLF) and 40 healthy subjects. SI was assessed by measuring 29 cytokines and the redox state of circulating albumin (HNA2), a marker of systemic oxidative stress. Systemic circulatory dysfunction (SCD) was estimated by plasma renin (PRC) and copeptin (PCC) concentrations. Measurements were performed at enrollment (baseline) in all patients and sequentially during hospitalization in 255. The main findings of this study were: (1) Patients with AD without ACLF showed very high baseline levels of inflammatory cytokines, HNA2, PRC, and PCC. Patients with ACLF showed significantly higher levels of these markers than those without ACLF; (2) different cytokine profiles were identified according to the type of ACLF precipitating event (active alcoholism/acute alcoholic hepatitis, bacterial infection, and others); (3) severity of SI and frequency and severity of ACLF at enrollment were strongly associated. The course of SI and the course of ACLF (improvement, no change, or worsening) during hospitalization and short‐term mortality were also strongly associated; and (4) the strength of association of ACLF with SI was higher than with SCD. Conclusion: These data support SI as the primary driver of ACLF in cirrhosis. (Hepatology 2016;64:1249‐1264).


Journal of Thrombosis and Haemostasis | 2005

Transplantation of endothelial cells corrects the phenotype in hemophilia A mice

Vinay Kumaran; Daniel Benten; Antonia Follenzi; Brigid Joseph; Rita Sarkar; Sanjeev Gupta

Summary.  Background: The deficiency of factor VIII, a co‐factor in the intrinsic coagulation pathway results in hemophilia A. Although FVIII is synthesized largely in the liver, the specific liver cell type(s) responsible for FVIII production is controversial. Objective: This study aimed to determine the cellular origin of FVIII synthesis and release in mouse models. Methods: We transplanted cells into the peritoneal cavity of hemophilia A knockout mice. Plasma FVIII activity was measured using a Chromogenix assay 2–7 days after cell transplantation, and phenotypic correction was determined with tail‐clip challenge 7 days following cell transplantation. Transplanted cells were identified by histologic and molecular assays. Results: Untreated hemophilia A mice, as well as mice treated with the hepatocyte‐enriched fraction, showed extensive mortality following tail‐clip challenge. In contrast, recipients of unfractionated liver cells (mixture of hepatocytes, liver sinusoidal endothelial cells (LSEC), Kupffer cells, and hepatic stellate cells) or of the cell fraction enriched in LSECs survived tail‐clip challenge (P < 0.001). FVIII was secreted in the blood stream in recipients of unfractionated liver cells, LSECs and pancreatic islet‐derived MILE SVEN 1 (MS1) endothelial cells. Although transplanted hepatocytes maintained functional integrity in the peritoneal cavity, these cells did not produce detectable plasma FVIII activity. Conclusions: The assay of cell transplantation in the peritoneal cavity showed that endothelial cells but not hepatocytes produced phenotypic correction in hemophilia A mice. Therefore, endothelial cells should be suitable additional targets for cell and gene therapy in hemophilia A.


Hepatology | 2005

Hepatocyte transplantation activates hepatic stellate cells with beneficial modulation of cell engraftment in the rat

Daniel Benten; Vinay Kumaran; Brigid Joseph; Jörn M. Schattenberg; Yury Popov; Detlef Schuppan; Sanjeev Gupta

We investigated whether transplanted hepatocytes interact with hepatic stellate cells, as cell–cell interactions could modulate their engraftment in the liver. We transplanted Fischer 344 rat hepatocytes into syngeneic dipeptidyl peptidase IV–deficient rats. Activation of hepatic stellate cells was analyzed by changes in gene expression, including desmin and α‐smooth muscle actin, matrix proteases and their inhibitors, growth factors, and other stellate cell‐associated genes with histological methods or polymerase chain reaction. Furthermore, the potential role of hepatic ischemia, Kupffer cells, and cytokine release in hepatic stellate cell activation was investigated. Hepatocyte transplantation activated desmin‐positive hepatic stellate cells, as well as Kupffer cells, including in proximity with transplanted cells. Inhibition of Kupffer cells by gadolinium chloride, blockade of tumor necrosis factor alpha (TNF‐α) activity with etanercept or attenuation of liver ischemia with nitroglycerin did not decrease this hepatic stellate cell perturbation. After cell transplantation, soluble signals capable of activating hepatic stellate cells were rapidly induced, along with early upregulated expression of matrix metalloproteinases‐2, ‐3, ‐9, ‐13, ‐14, and their inhibitors. Moreover, prior depletion of activated hepatic stellate cells with gliotoxin decreased transplanted cell engraftment. In conclusion, cell transplantation activated hepatic stellate cells, which, in turn, contributed to transplanted cell engraftment in the liver. Manipulation of hepatic stellate cells might provide new strategies to improve liver repopulation after enhanced transplanted cell engraftment. Supplementary material for this article can be found on the HEPATOLOGY website (http://interscience.wiley.com/jpages/0270‐9139/suppmat/index.html). (HEPATOLOGY 2005;42:1072–1081.)


Nature Protocols | 2012

RGB marking with lentiviral vectors for multicolor clonal cell tracking

Kristoffer Weber; Michael Thomaschewski; Daniel Benten; Boris Fehse

Cells transduced with lentiviral vectors are individually marked by a highly characteristic pattern of insertion sites inherited by all their progeny. We have recently extended this principle of clonal cell marking by introducing the method of RGB marking, which makes use of the simultaneous transduction of target cells with three lentiviral gene ontology (LeGO) vectors encoding red, green or blue fluorescent proteins. In accordance with the additive color model, individual RGB-marked cells display a large variety of unique and highly specific colors. Color codes remain stable after cell division and can thus be used for clonal tracking in vivo and in vitro. Our protocol for efficient RGB marking is based on established methods of lentiviral vector production (3–4 d) and titration (3 d). The final RGB-marking step requires concurrent transduction with the three RGB vectors at equalized multiplicities of infection (1–12 h). The initial efficiency of RGB marking can be assessed after 2–4 d by flow cytometry and/or fluorescence microscopy.


Hepatology | 2005

Hepatic targeting of transplanted liver sinusoidal endothelial cells in intact mice

Daniel Benten; Antonia Follenzi; Kuldeep K. Bhargava; Vinay Kumaran; Christopher J. Palestro; Sanjeev Gupta

Targeting of cells to specific tissues is critical for cell therapy. To study endothelial cell targeting, we isolated mouse liver sinusoidal endothelial cells (LSEC) and examined cell biodistributions in animals. To identify transplanted LSEC in tissues, we labeled cells metabolically with DiI‐conjugated acetylated low density lipoprotein particles (DiI‐Ac‐LDL) or 111Indium‐oxine, used LSEC from Rosa26 donors expressing β‐galactosidase or Tie‐2‐GFP donors with green fluorescent protein (GFP) expression, and tranduced LSEC with a GFP‐lentiviral vector. LSEC efficiently incorporated 111Indium and DiI‐Ac‐LDL and expressed GFP introduced by the lentiviral vector. Use of radiolabeled LSEC showed differences in cell biodistributions in relation to the cell transplantation route. After intraportal injection, LSEC were largely in the liver (60 ± 13%) and, after systemic intravenous injection, in lungs (67 ± 9%); however, after intrasplenic injection, only some LSEC remained in the spleen (29 ± 10%; P < .01), whereas most LSEC migrated to the liver or lungs. Transplanted LSEC were found in the liver, lungs, and spleen shortly after transplantation, whereas longer‐term cell survival was observed only in the liver. Transplanted LSEC were distinct from Kupffer cells with expression of Tie‐2 promoter‐driven GFP and of CD31, without F4/80 reactivity. In further studies using radiolabeled LSEC, we established that the manipulation of receptor‐mediated cell adhesion in liver sinusoids or the manipulation of blood flow–dependent cell exit from sinusoids improved intrahepatic retention of LSEC to 89 ± 7% and 89 ± 5%, respectively (P < .01). In conclusion, the targeting of LSEC to the liver and other organs is directed by vascular bed–specific mechanisms, including blood flow–related processes, and cell‐specific factors. These findings may facilitate analysis of LSEC for cell and gene therapy applications. (HEPATOLOGY 2005.)

Collaboration


Dive into the Daniel Benten's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sanjeev Gupta

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Brigid Joseph

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Vinay Kumaran

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

T. Volz

University of Hamburg

View shared research outputs
Top Co-Authors

Avatar

Antonia Follenzi

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge