Network


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

Hotspot


Dive into the research topics where Asha Balakrishnan is active.

Publication


Featured researches published by Asha Balakrishnan.


Hepatology | 2013

MicroRNA‐221 overexpression accelerates hepatocyte proliferation during liver regeneration

Qinggong Yuan; Komal Loya; Bhavna Rani; Selina Möbus; Asha Balakrishnan; Jutta Lamlé; Toni Cathomen; Arndt Vogel; Michael P. Manns; Michael Ott; Tobias Cantz; Amar Deep Sharma

The tightly controlled replication of hepatocytes in liver regeneration and uncontrolled proliferation of tumor cells in hepatocellular carcinoma (HCC) are often modulated by common regulatory pathways. Several microRNAs (miRNAs) are involved in HCC progression by modulating posttranscriptional expression of multiple target genes. miR‐221, which is frequently up‐regulated in HCCs, delays fulminant liver failure in mice by inhibiting apoptosis, indicating a pleiotropic role of miR‐221 in hepatocytes. Here, we hypothesize that modulation of miR‐221 targets in primary hepatocytes enhances proliferation, providing novel clues for enhanced liver regeneration. We demonstrate that miR‐221 enhances proliferation of in vitro cultivated primary hepatocytes. Furthermore, applying two‐thirds partial hepatectomy as a surgically induced liver regeneration model we show that adeno‐associated virus‐mediated overexpression of miR‐221 in the mouse liver also accelerates hepatocyte proliferation in vivo. miR‐221 overexpression leads to rapid S‐phase entry of hepatocytes during liver regeneration. In addition to the known targets p27 and p57, we identify Aryl hydrocarbon nuclear translocator (Arnt) messenger RNA (mRNA) as a novel target of miR‐221, which contributes to the pro‐proliferative activity of miR‐221. Conclusion: miR‐221 overexpression accelerates hepatocyte proliferation. Pharmacological intervention targeting miR‐221 may thus be therapeutically beneficial in liver failure by preventing apoptosis and by inducing liver regeneration. (HEPATOLOGY 2013;)


Journal of Hepatology | 2015

MicroRNA-199a-5p inhibition enhances the liver repopulation ability of human embryonic stem cell-derived hepatic cells

Selina Möbus; Dakai Yang; Qinggong Yuan; Timo H.-W. Lüdtke; Asha Balakrishnan; Malte Sgodda; Bhavna Rani; Andreas Kispert; Marcos J. Araúzo-Bravo; Arndt Vogel; Michael P. Manns; Michael Ott; Tobias Cantz; Amar Deep Sharma

BACKGROUND & AIMS Current hepatic differentiation protocols for human embryonic stem cells (ESCs) require substantial improvements. MicroRNAs (miRNAs) have been reported to regulate hepatocyte cell fate during liver development, but their utility to improve hepatocyte differentiation from ESCs remains to be investigated. Therefore, our aim was to identify and to analyse hepatogenic miRNAs for their potential to improve hepatocyte differentiation from ESCs. METHODS By miRNA profiling and in vitro screening, we identified miR-199a-5p among several potential hepatogenic miRNAs. Transplantation studies of miR-199a-5p-inhibited hepatocyte-like cells (HLCs) in the liver of immunodeficient fumarylacetoacetate hydrolase knockout mice (Fah(-/-)/Rag2(-/-)/Il2rg(-/-)) were performed to assess their in vivo liver repopulation potential. For target determination, western blot and luciferase reporter assay were carried out. RESULTS miRNA profiling revealed 20 conserved candidate hepatogenic miRNAs. By miRNA screening, only miR-199a-5p inhibition in HLCs was found to be able to enhance the in vitro hepatic differentiation of mouse as well as human ESCs. miR-199a-5p inhibition in human ESCs-derived HLCs enhanced their engraftment and repopulation capacity in the liver of Fah(-/-)/Rag2(-/-)/Il2rg(-/-) mice. Furthermore, we identified SMARCA4 and MST1 as novel targets of miR-199a-5p that may contribute to the improved hepatocyte generation and in vivo liver repopulation. CONCLUSIONS Our findings demonstrate that miR-199a-5p inhibition in ES-derived HLCs leads to improved hepatocyte differentiation. Upon transplantation, HLCs were able to engraft and repopulate the liver of Fah(-/-)/Rag2(-/-)/Il2rg(-/-) mice. Thus, our findings suggest that miRNA modulation may serve as a promising approach to generate more mature HLCs from stem cell sources for the treatment of liver diseases.


Genes, Chromosomes and Cancer | 2006

Quantitative microsatellite analysis to delineate the commonly deleted region 1p22.3 in mantle cell lymphomas

Asha Balakrishnan; Nils von Neuhoff; Cornelia Rudolph; Kathrin Kamphues; Margit Schraders; Patricia J. T. A. Groenen; Johan H. J. M. van Krieken; Evelyne Callet-Bauchu; Brigitte Schlegelberger; Doris Steinemann

The molecular pathogenesis of mantle cell lymphomas (MCL), a subset of B‐cell non‐Hodgkins lymphomas with a poor prognosis, is still poorly understood. In addition to the characteristic primary genetic alteration t(11;14)(q13;q32), several further genetic changes are present in most cases. One of the most frequent genomic imbalances is the deletion of 1p22.1–p31.1 observed in nearly one‐third of MCL cases. This might indicate the presence of tumor suppressor gene(s) in this critical region of deletion. Quantitative microsatellite analysis (QuMA) is a real‐time PCR‐based method to detect DNA copy number changes. Since QuMA has the resolving power to detect subtle genomic alterations, including homozygous deletions, this may help to identify candidate tumor suppressor genes from deleted regions. To gain more insight into the molecular pathogenesis of MCL, QuMA was performed on genomic DNA from 57 MCL cases. Eight microsatellite loci mapping to the chromosomal region 1p22.3 were analyzed. Losses were observed in 51 of the 57 (∼89.5%) samples. Two cases showed a homozygous deletion at the locus containing the gene SH3GLB1, which plays a key role in Bax‐mediated apoptosis. Two hotspots with copy number losses were detected at chromosomal localizations 85.4 and 86.6 Mb encompassing BCL10 and CLCA2. Both the genes seem to be attractive candidates to study tumor suppressor function in MCL. This article contains Supplementary material available at http://www.interscience.wiley.com/jpages/1045–2257/suppmat.


Nature Communications | 2016

MicroRNA-125b-5p mimic inhibits acute liver failure

Dakai Yang; Qinggong Yuan; Asha Balakrishnan; Heike Bantel; Jan-Henning Klusmann; Michael P. Manns; Michael Ott; Tobias Cantz; Amar Deep Sharma

The lack of broad-spectrum anti-acute liver failure (ALF) therapeutic agents contributes to ALF-related mortality. MicroRNAs (miRNAs) are suggested to be potent serum biomarkers for ALF, but their functional and therapeutic relevance in ALF are unclear. Here we show an unbiased approach, using two complementary miRNA screens, to identify miRNAs that can attenuate ALF. We identify miR-125b-5p as a regulator of cell death that attenuates paracetamol-induced and FAS-induced toxicity in mouse and human hepatocytes. Importantly, administration of miR-125b-5p mimic in mouse liver prevents injury and improves survival in models of ALF. Functional studies show that miR-125b-5p ameliorates ALF by directly regulating kelch-like ECH-associated protein 1, in turn elevating expression of nuclear factor-E2-related factor 2, a known regulator in ALF. Collectively, our findings establish miR-125b-5p as an important regulator of paracetamol-induced and FAS-induced cell death. Thus, miR-125b-5p mimic may serve as a broad-spectrum therapeutic attenuator of cell death during ALF.


Chemistry: A European Journal | 2017

Synthesis of Magnetic-Nanoparticle/Ansamitocin Conjugates—Inductive Heating Leads to Decreased Cell Proliferation In Vitro and Attenuation Of Tumour Growth In Vivo

Katja Seidel; Asha Balakrishnan; Christoph Alexiou; Christina Janko; Ronja-Melinda Komoll; Liang-Liang Wang; Andreas Kirschning; Michael Ott

Conjugates based on nanostructured, superparamagnetic particles, a thermolabile linker and a cytotoxic maytansinoid were developed to serve as a model for tumour-selective drug delivery and release. It combines chemo- with thermal therapy. The linker-modified toxin was prepared by a combination of biotechnology and semisynthesis. Drug release was achieved by hyperthermia through an external oscillating electromagnetic field that induces heat inside the particles. Efficacy of this release concept was demonstrated both for cancer cell proliferation in vitro, and for tumour growth in vivo, in a xenograft mouse model. Biocompatibility studies for these magnetic-nanoparticle/ansamitocin conjugates complement this work.


Chemistry: A European Journal | 2017

A Bio-Chemosynthetic Approach to Superparamagnetic Iron Oxide-Ansamitocin Conjugates for Use in Magnetic Drug Targeting

Liang-Liang Wang; Asha Balakrishnan; Nadja-Carola Bigall; David Candito; Jan F. Miethe; Katja Seidel; Yu Xie; Michael Ott; Andreas Kirschning

A combination of mutasynthesis using a mutant strain of A. pretiosum blocked in the biosynthesis of amino-hydroxybenzoic acid (AHBA) and semisynthesis relying on a Stille cross-coupling step provided access to new ansamitocin derivatives of which one was attached by a thermolabile linker to nanostructured iron oxide particles. When exposed to an oscillating electromagnetic field the resulting iron oxide/ansamitocin conjugate 19 heats up in an aqueous suspension and the ansamitocin derivative 16 is released by means of a retro-Diels-Alder reaction. It exerts strong antiproliferative activity (IC50 =4.8 ng mg-1 ) in mouse fibroblasts. These new types of conjugates have the potential for combating cancer through hyperthermia and chemotherapy using an electromagnetic external trigger.


World Journal of Hepatology | 2018

Homologous recombination mediates stable Fah gene integration and phenotypic correction in tyrosinaemia mouse-model

Qinggong Yuan; Thu Huong Vu; Simon Krooss; Christien Bednarski; Asha Balakrishnan; Toni Cathomen; Michael P. Manns; Ulrich Baumann; Amar Deep Sharma; Michael Ott

AIM To stably correct tyrosinaemia in proliferating livers of fumarylacetoacetate-hydrolase knockout (Fah-/-) mice by homologous-recombination-mediated targeted addition of the Fah gene. METHODS C57BL/6 Fah∆exon5 mice served as an animal model for human tyrosinaemia type 1 in our study. The vector was created by amplifying human Fah cDNA including the TTR promoter from a lentivirus plasmid as described. The Fah expression cassette was flanked by homologous arms (620 bp and 749 bp long) of the Rosa26 gene locus. Mice were injected with 2.1 × 108 VP of this vector (rAAV8-ROSA26.HAL-TTR.Fah-ROSA26.HAR) via the tail vein. Mice in the control group were injected with 2.1 × 108 VP of a similar vector but missing the homologous arms (rAAV8-TTR.Fah). Primary hepatocytes from Fah-/- recipient mice, treated with our vectors, were isolated and 1 × 106 hepatocytes were transplanted into secondary Fah-/- recipient mice by injection into the spleen. Upon either vector application or hepatocyte transplantation NTBC treatment was stopped in recipient mice. RESULTS Here, we report successful HR-mediated genome editing by integration of a Fah gene expression cassette into the “safe harbour locus” Rosa26 by recombinant AAV8. Both groups of mice showed long-term survival, weight gain and FAH positive clusters as determined by immunohistochemistry analysis of liver sections in the absence of NTBC treatment. In the group of C57BL/6 Fah∆exon5 mice, which have been transplanted with hepatocytes from a mouse injected with rAAV8-ROSA26.HAL-TTR.Fah-ROSA26.HAR 156 d before, 6 out of 6 mice showed long-term survival, weight gain and FAH positive clusters without need for NTBC treatment. In contrast only 1 out 5 mice, who received hepatocytes from rAAV8-TTR.Fah treated mice, survived and showed few and smaller FAH positive clusters. These results demonstrate that homologous recombination-mediated Fah gene transfer corrects the phenotype in a mouse model of human tyrosinaemia type 1 (Fah-/- mice) and is long lasting in a proliferating state of the liver as shown by withdrawal of NTBC treatment and serial transplantation of isolated hepatocytes from primary Fah-/- recipient mice into secondary Fah-/- recipient mice. This long term therapeutic efficacy is clearly superior to our control mice treated with episomal rAAV8 gene therapy approach. CONCLUSION HR-mediated rAAV8 gene therapy provides targeted transgene integration and phenotypic correction in Fah-/- mice with superior long-term efficacy compared to episomal rAAV8 therapy in proliferating livers.


Cell Death and Disease | 2018

Let-7c inhibits cholangiocarcinoma growth but promotes tumor cell invasion and growth at extrahepatic sites

Yu Xie; Hang Zhang; Xingjun Guo; Ye-Chen Feng; Ruizhi He; Xu Li; Shuo Yu; Yan Zhao; Ming Shen; Feng Zhu; Xin Wang; Min Wang; Asha Balakrishnan; Michael Ott; Feng Peng; Renyi Qin

Cholangiocarcinoma (CCA) is a cancer type with high postoperative relapse rates and poor long-term survival largely due to tumor invasion, distant metastasis, and multidrug resistance. Deregulated microRNAs (miRNAs) are implicated in several cancer types including CCA. The specific roles of the miRNA let-7c in cholangiocarcinoma are not known and need to be further elucidated. In our translational study we show that microRNA let-7c expression was significantly downregulated in human cholangiocarcinoma tissues when compared to adjacent tissues of the same patient. Let-7c inhibited the tumorigenic properties of cholangiocarcinoma cells including their self-renewal capacity and sphere formation in vitro and subcutaneous cancer cell growth in vivo. Ectopic let-7c overexpression suppressed migration and invasion capacities of cholangiocarcinoma cell lines in vitro, however, promoted distant invasiveness in vivo. Furthermore, we found that let-7c regulated the aforementioned malignant biological properties, at least in part, through regulation of EZH2 protein expression and through the DVL3/β-catenin axis. The miRNA let-7c thus plays an important dual role in regulating tumorigenic and metastatic abilities of human cholangiocarcinoma through mechanisms involving EZH2 protein and the DVL3/β-catenin axis.


MicroRNA in Regenerative Medicine | 2015

Chapter 36 – MicroRNAs in Liver Regeneration

Dakai Yang; Selina Möbus; Asha Balakrishnan; Amar Deep Sharma

The liver is an organ with multiple unique functions and a high regenerative capacity, able to regain its size, function, and structure from as little as 10% of the residual mass. The restoration of liver mass is accomplished in initiation, proliferation, and termination phases, which are regulated by a complex network of growth factors and cytokines. MicroRNAs (miRNAs) have emerged as important mediators of gene regulation. Significant changes in miRNA expression occur during two-thirds partial hepatectomy, which represents a well-established in vivo model for liver regeneration. This chapter summarizes the miRNAs involved in liver regeneration reported to date, including their function in liver homeostasis and their therapeutic utility in treatment of end-stage liver diseases.


Cell Stem Cell | 2016

Direct Reprogramming of Hepatic Myofibroblasts into Hepatocytes In Vivo Attenuates Liver Fibrosis

Guangqi Song; Martin Pacher; Asha Balakrishnan; Qinggong Yuan; Hsin-Chieh Tsay; Dakai Yang; Julia Reetz; Sabine Brandes; Zhen Dai; Brigitte M. Pützer; Marcos J. Araúzo-Bravo; Doris Steinemann; Tom Luedde; Robert F. Schwabe; Michael P. Manns; Hans R. Schöler; Axel Schambach; Tobias Cantz; Michael Ott; Amar Deep Sharma

Collaboration


Dive into the Asha Balakrishnan's collaboration.

Top Co-Authors

Avatar

Michael Ott

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dakai Yang

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Tobias Cantz

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arndt Vogel

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Bhavna Rani

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge