Network


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

Hotspot


Dive into the research topics where Maike Zimmermann is active.

Publication


Featured researches published by Maike Zimmermann.


Biochemistry | 2010

Targeting of Protein Phosphatases PP2A and PP2B to the C-terminus of the L-type Calcium Channel Cav1.2

Hui Xu; Kenneth S. Ginsburg; Duane D. Hall; Maike Zimmermann; Ivar S. Stein; Mingxu Zhang; Samvit Tandan; Joseph A. Hill; Mary C. Horne; Donald M. Bers; Johannes W. Hell

The L-type Ca(2+) channel Ca(v)1.2 forms macromolecular signaling complexes that comprise the β(2) adrenergic receptor, trimeric G(s) protein, adenylyl cyclase, and cAMP-dependent protein kinase (PKA) for efficient signaling in heart and brain. The protein phosphatases PP2A and PP2B are part of this complex. PP2A counteracts increase in Ca(v)1.2 channel activity by PKA and other protein kinases, whereas PP2B can either augment or decrease Ca(v)1.2 currents in cardiomyocytes depending on the precise experimental conditions. We found that PP2A binds to two regions in the C-terminus of the central, pore-forming α(1) subunit of Ca(v)1.2: one region spans residues 1795-1818 and the other residues 1965-1971. PP2B binds immediately downstream of residue 1971. Injection of a peptide that contained residues 1965-1971 and displaced PP2A but not PP2B from endogenous Ca(v)1.2 increased basal and isoproterenol-stimulated L-type Ca(2+) currents in acutely isolated cardiomyocytes. Together with our biochemical data, these physiological results indicate that anchoring of PP2A at this site of Ca(v)1.2 in the heart negatively regulates cardiac L-type currents, likely by counterbalancing basal and stimulated phosphorylation that is mediated by PKA and possibly other kinases.


Cell Cycle | 2016

Cyclin G2 promotes cell cycle arrest in breast cancer cells responding to fulvestrant and metformin and correlates with patient survival

Maike Zimmermann; Aruni S. Arachchige-Don; Michaela S. Donaldson; Tommaso Patriarchi; Mary C. Horne

ABSTRACT Definition of cell cycle control proteins that modify tumor cell resistance to estrogen (E2) signaling antagonists could inform clinical choice for estrogen receptor positive (ER+) breast cancer (BC) therapy. Cyclin G2 (CycG2) is upregulated during cell cycle arrest responses to cellular stresses and growth inhibitory signals and its gene, CCNG2, is directly repressed by E2-bound ER complexes. Our previous studies showed that blockade of HER2, PI3K and mTOR signaling upregulates CycG2 expression in HER2+ BC cells, and that CycG2 overexpression induces cell cycle arrest. Moreover, insulin and insulin-like growth factor-1 (IGF-1) receptor signaling strongly represses CycG2. Here we show that blockade of ER-signaling in MCF7 and T47D BC cell lines enhances the expression and nuclear localization of CycG2. Knockdown of CycG2 attenuated the cell cycle arrest response of E2-depleted and fulvestrant treated MCF7 cells. These muted responses were accompanied by sustained inhibitory phosphorylation of retinoblastoma (RB) protein, expression of cyclin D1, phospho-activation of ERK1/2 and MEK1/2 and expression of cRaf. Our work indicates that CycG2 can form complexes with CDK10, a CDK linked to modulation of RAF/MEK/MAPK signaling and tamoxifen resistance. We determined that metformin upregulates CycG2 and potentiates fulvestrant-induced CycG2 expression and cell cycle arrest. CycG2 knockdown blunts the enhanced anti-proliferative effect of metformin on fulvestrant treated cells. Meta-analysis of BC tumor microarrays indicates that CCNG2 expression is low in aggressive, poor-prognosis BC and that high CCNG2 expression correlates with longer periods of patient survival. Together these findings indicate that CycG2 contributes to signaling networks that limit BC.


Biochemical Journal | 2014

Down-regulation of cyclin G2 by insulin, IGF-I (insulin-like growth factor 1) and X10 (AspB10 insulin): Role in mitogenesis

Angela Manegold Svendsen; Sofia B. Winge; Maike Zimmermann; Anne B. Lindvig; Caroline B. Warzecha; Waseem Sajid; Mary C. Horne; Pierre De Meyts

The mechanisms whereby insulin analogues may cause enhanced mitogenicity through activation of either the IR (insulin receptor) or the IGF-IR (insulin-like growth factor 1 receptor) are incompletely understood. We demonstrate that in L6 myoblasts expressing only IGF-IRs as well as in the same cells overexpressing the IR, IGF-I (insulin-like growth factor 1), insulin and X10 (AspB10 insulin) down-regulate the mRNA expression level of the cell cycle inhibitor cyclin G2, as measured by qRT-PCR (quantitative reverse transcription-PCR), and induce cell growth measured by [6-(3)H]thymidine incorporation into DNA. Western blotting showed a marked down-regulation of cyclin G2 at the protein level in both cell lines. Overexpression of cyclin G2 in the two cell lines diminished the mitogenic effect of all three ligands. The use of specific inhibitors indicated that both the MAPK (mitogen-activated protein kinase) and the PI3K (phosphoinositide 3-kinase) pathways mediate the down-regulation of Ccng2. The down-regulation of CCNG2 by the three ligands was also observed in other cell lines: MCF-7, HMEC, Saos-2, R(-)/IR and INS-1. These results indicate that regulation of cyclin G2 is a key mechanism whereby insulin, insulin analogues and IGF-I stimulate cell proliferation.


Chemical Research in Toxicology | 2017

DNA Adducts from Anticancer Drugs as Candidate Predictive Markers for Precision Medicine

Alessia Stornetta; Maike Zimmermann; George D. Cimino; Paul T. Henderson; Shana J. Sturla

Biomarker-driven drug selection plays a central role in cancer drug discovery and development, and in diagnostic strategies to improve the use of traditional chemotherapeutic drugs. DNA-modifying anticancer drugs are still used as first line medication, but drawbacks such as resistance and side effects remain an issue. Monitoring the formation and level of DNA modifications induced by anticancer drugs is a potential strategy for stratifying patients and predicting drug efficacy. In this perspective, preclinical and clinical data concerning the relationship between drug-induced DNA adducts and biological response for platinum drugs and combination therapies, nitrogen mustards and half-mustards, hypoxia-activated drugs, reductase-activated drugs, and minor groove binding agents are presented and discussed. Aspects including measurement strategies, identification of adducts, and biological factors that influence the predictive relationship between DNA modification and biological response are addressed. A positive correlation between DNA adduct levels and response was observed for the majority of the studies, demonstrating the high potential of using DNA adducts from anticancer drugs as mechanism-based biomarkers of susceptibility, especially as bioanalysis approaches with higher sensitivity and throughput emerge.


Molecular Cancer Therapeutics | 2017

Microdose-induced Drug-DNA Adducts as Biomarkers of Chemotherapy Resistance in Humans and Mice

Maike Zimmermann; Si Si Wang; Hongyong Zhang; Tzu Yin Lin; Michael A. Malfatti; Kurt W. Haack; Ted Ognibene; Hongyuan Yang; Susan D. Airhart; Kenneth W. Turteltaub; George D. Cimino; Clifford G. Tepper; Alexandra Drakaki; Karim Chamie; Ralph de Vere White; Chong Xian Pan; Paul T. Henderson

We report progress on predicting tumor response to platinum-based chemotherapy with a novel mass spectrometry approach. Fourteen bladder cancer patients were administered one diagnostic microdose each of [14C]carboplatin (1% of the therapeutic dose). Carboplatin–DNA adducts were quantified by accelerator mass spectrometry in blood and tumor samples collected within 24 hours, and compared with subsequent chemotherapy response. Patients with the highest adduct levels were responders, but not all responders had high adduct levels. Four patient-derived bladder cancer xenograft mouse models were used to test the possibility that another drug in the regimen could cause a response. The mice were dosed with [14C]carboplatin or [14C]gemcitabine and the resulting drug–DNA adduct levels were compared with tumor response to chemotherapy. At least one of the drugs had to induce high drug–DNA adduct levels or create a synergistic increase in overall adducts to prompt a corresponding therapeutic response, demonstrating proof-of-principle for drug–DNA adducts as predictive biomarkers. Mol Cancer Ther; 16(2); 376–87. ©2016 AACR.


PLOS ONE | 2016

Molecular Dissection of Induced Platinum Resistance through Functional and Gene Expression Analysis in a Cell Culture Model of Bladder Cancer

Sisi Wang; Hongyong Zhang; Tiffany M. Scharadin; Maike Zimmermann; Bin Hu; Amy Wang Pan; Ruth L. Vinall; Tzu Yin Lin; George D. Cimino; Patrick Chain; Momchilo Vuyisich; Cheryl D. Gleasner; Kim McMurry; Michael A. Malfatti; Kenneth W. Turteltaub; Ralph de Vere White; Chong Xian Pan; Paul T. Henderson

We report herein the development, functional and molecular characterization of an isogenic, paired bladder cancer cell culture model system for studying platinum drug resistance. The 5637 human bladder cancer cell line was cultured over ten months with stepwise increases in oxaliplatin concentration to generate a drug resistant 5637R sub cell line. The MTT assay was used to measure the cytotoxicity of several bladder cancer drugs. Liquid scintillation counting allowed quantification of cellular drug uptake and efflux of radiolabeled oxaliplatin and carboplatin. The impact of intracellular drug inactivation was assessed by chemical modulation of glutathione levels. Oxaliplatin- and carboplatin-DNA adduct formation and repair was measured using accelerator mass spectrometry. Resistance factors including apoptosis, growth factor signaling and others were assessed with RNAseq of both cell lines and included confirmation of selected transcripts by RT-PCR. Oxaliplatin, carboplatin, cisplatin and gemcitabine were significantly less cytotoxic to 5637R cells compared to the 5637 cells. In contrast, doxorubicin, methotrexate and vinblastine had no cell line dependent difference in cytotoxicity. Upon exposure to therapeutically relevant doses of oxaliplatin, 5637R cells had lower drug-DNA adduct levels than 5637 cells. This difference was partially accounted for by pre-DNA damage mechanisms such as drug uptake and intracellular inactivation by glutathione, as well as faster oxaliplatin-DNA adduct repair. In contrast, both cell lines had no significant differences in carboplatin cell uptake, efflux and drug-DNA adduct formation and repair, suggesting distinct resistance mechanisms for these two closely related drugs. The functional studies were augmented by RNAseq analysis, which demonstrated a significant change in expression of 83 transcripts, including 50 known genes and 22 novel transcripts. Most of the transcripts were not previously associated with bladder cancer chemoresistance. This model system and the associated phenotypic and genotypic data has the potential to identify some novel details of resistance mechanisms of clinical importance to bladder cancer.


Molecular Cancer Therapeutics | 2017

COX-2/sEH Dual Inhibitor PTUPB Potentiates the Antitumor Efficacy of Cisplatin

Fuli Wang; Hongyong Zhang; Ai Hong Ma; Weimin Yu; Maike Zimmermann; Jun Yang; Sung Hee Hwang; Daniel Zhu; Tzu Yin Lin; Michael A. Malfatti; Kenneth W. Turteltaub; Paul T. Henderson; Susan D. Airhart; Bruce D. Hammock; Jianlin Yuan; Ralph W. deVere White; Chong Xian Pan

Cisplatin-based therapy is highly toxic, but moderately effective in most cancers. Concurrent inhibition of cyclooxygenase-2 (COX-2) and soluble epoxide hydrolase (sEH) results in antitumor activity and has organ-protective effects. The goal of this study was to determine the antitumor activity of PTUPB, an orally bioavailable COX-2/sEH dual inhibitor, in combination with cisplatin and gemcitabine (GC) therapy. NSG mice bearing bladder cancer patient-derived xenografts were treated with vehicle, PTUPB, cisplatin, GC, or combinations thereof. Mouse experiments were performed with two different PDX models. PTUPB potentiated cisplatin and GC therapy, resulting in significantly reduced tumor growth and prolonged survival. PTUPB plus cisplatin was no more toxic than cisplatin single-agent treatment as assessed by body weight, histochemical staining of major organs, blood counts, and chemistry. The combination of PTUPB and cisplatin increased apoptosis and decreased phosphorylation in the MAPK/ERK and PI3K/AKT/mTOR pathways compared with controls. PTUPB treatment did not alter platinum–DNA adduct levels, which is the most critical step in platinum-induced cell death. The in vitro study using the combination index method showed modest synergy between PTUPB and platinum agents only in 5637 cell line among several cell lines examined. However, PTUPB is very active in vivo by inhibiting angiogenesis. In conclusion, PTUPB potentiated the antitumor activity of cisplatin-based treatment without increasing toxicity in vivo and has potential for further development as a combination chemotherapy partner. Mol Cancer Ther; 17(2); 474–83. ©2017 AACR.


Journal of Clinical Oncology | 2015

A microdosing study to identify chemoresistance in bladder cancer.

Helen Chow; Sisi Wang; Hongyong Zhang; Maike Zimmermann; Tzu-yin Lin; Tiffany M. Scharadin; Michael A. Malfatti; Christopher P. Evans; George D. Cimino; Kenneth W. Turteltaub; Ralph de Vere White; Paul T. Henderson; Chong Xian Pan

356 Background: DNA adduct formation and incorporation of gemcitabine into genomic DNA are critical steps in cancer cell response to platinum (Pt) and gemcitabine chemotherapy, respectively. We hypothesize that levels of Pt-DNA adducts and gemcitabine in genomic DNA below a threshold are predictive of chemoresistance. Accelerator mass spectrometry (AMS) is an ultrasensitive method for measuring radiocarbon. By measuring 14C bound to DNA, AMS was used to quantify carboplatin-DNA damage and gemcitabine incorporation into DNA after mice or patients received nontoxic “microdoses” of 14C-labeled carboplatin or gemcitabine. Methods: Cancer cells and mice bearing tumor xenografts were treated with one microdose (1% of the therapeutic dose) or therapeutic dose of [14C]carboplatin or [14C]gemcitabine. Carboplatin-DNA adducts and gemcitabine incorporation in DNA were correlated with cell/tumor response to chemotherapy. In the Phase 0 trial, patients with advanced bladder or non-small cell lung cancer were treated w...


Cancer Research | 2014

Abstract 2821: Dual-label microdosing approach to identify chemoresistance to carboplatin and gemcitabine combination chemotherapy

Maike Zimmermann; Tiffany M. Scharadin; Hongyong Zhang; Tzu-yin Lin; Ralph W. deVere White; Chong Xian Pan; Paul T. Henderson

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Gemcitabine is commonly given in combination with cisplatin or carboplatin (GC therapy) as the first-line chemotherapy regimen for several malignancies, including bladder cancer. Unfortunately, less than half of patients respond to the therapy, generating a critical need for a test that predicts tumor response to GC therapy. The goal of this project is to measure drug-DNA adducts after administration of a single non-toxic microdose of gemcitabine in combination with carboplatin and correlating the degree of modification to chemotherapy sensitivity. We have previously shown that resistance to carboplatin may be identified by using a [14C]carboplatin microdose (1% of therapeutic dose), and subsequent measurement of the [14C]-DNA adducts via accelerator mass spectrometry (AMS), an ultrasensitive method used for detecting rare isotopes. We postulate that adduct levels induced in tumors by GC microdoses are proportional to those formed during full dose chemotherapy. Furthermore, we hypothesize that there exists a threshold level of adducts above which tumors respond to GC therapy. We used bladder cancer cell lines and the nod scid gamma severe combined immunodeficient (NSG) mice containing patient-derived tumor xenografts (PDX) from bladder cancer patients to determine GC-DNA adduct levels using AMS. The NSG mouse is uniquely capable of accepting and propagating tumor tissue engrafted directly from patient biopsy samples. The resulting groups of mice each have identical tumors, but can be treated in a variety of ways with replicate experiments; characteristics that are impossible to achieve in normal clinical studies. We will report progress on developing protocols and preliminary GC microdosing data using the NSG-PDX mouse model of human bladder cancer. Citation Format: Maike Zimmermann, Tiffany M. Scharadin, Hongyong Zhang, Tzu-yin Lin, Ralph W. deVere White, Chong-xian Pan, Paul T. Henderson. Dual-label microdosing approach to identify chemoresistance to carboplatin and gemcitabine combination chemotherapy. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2821. doi:10.1158/1538-7445.AM2014-2821


Cancer Research | 2014

Abstract 905: Molecular dissection of platinum resistance through functional analysis

Amy Wang Pan; Sisi Wang; Hongyong Zhang; Ruth L. Vinall; Tzu-yin Lin; Michael A. Malfatti; Maike Zimmermann; Tiffany M. Scharadin; Kenneth W. Turteltaub; Ralph de Vere White; Chong Xian Pan; Paul T. Henderson

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA BACKGROUND: Platinum (Pt) agents (cisplatin, carboplatin and oxaliplatin) are active in many cancers including bladder cancer. Chemoresistance is the most common cause of treatment failure. This study is to determine the feasibility of using ultrasensitive accelerator mass spectrometry (AMS) to identify chemoresistance after cancer cells or patients are treated with one non-toxic microdose (1/100th of therapeutic dose) of Pt agents. The long-term goal is to identify chemoresistance before cancer patients receive toxic chemotherapy, and to determine the underlying resistance mechanisms to design personalized chemotherapy. METHODS: Cellular sensitivity to chemotherapeutic agents was determined by the MTT assay. Platinum-induced DNA adduct formation and repair of adducts was measured with AMS after cells were exposed 14C-labeled carboplatin and oxaliplatin. AMS quantifies the 14C label that is attached to genomic DNA when the 14C-labled drug forms adducts with DNA. Cell uptake and efflux was measured by liquid scintillation counting. Intracellular glutathione levels were measured by colorimetric analysis. RESULTS: Compared to the parental bladder cancer 5637 cells, chemoresistant 5637R cells are resistant to oxaliplatin (IC50: 2.45 µM versus 27.27 µM, p<0.0001), and cisplatin (0.59 µM versus 2.99 µM, p=0.049), carboplatin (24.34 µM versus 72.18 µM, p<0.0001), and gemcitabine (0.12 µM versus 1.44 µM, p=0.0015). Both 5637 and 5637R cells are still sensitive to other chemotherapeutic agents commonly used in treating bladder cancer, such as doxorubicin, methotrexate and vinblastine. Consistent with our hypothesis, chemoresistant 5637R cells have low oxaliplatin-induced DNA adduct levels than the parental 5637 cells (AUC of 943 versus 2,772 adducts per 109 nucleotide-hour for 5637, p=0.001). This low level of oxaliplatin-DNA adduct formation might be secondary to the pre-DNA damage mechanisms, such as decreased uptake (AUC of 4.42 versus 5.12 X 109 oxaliplatin molecules per cell for 5637, p=0.037) and increased intracellular inactivation of oxaliplatin by glutathione (53.91 versus 46.93 nmol/mg protein for 5637, p=0.003), plus increased repair of oxaliplatin-DNA adducts (3.48 versus 1.34 adducts per 108 nucleotides per hour for 5637, p=0.0004). We found the same correlation of low Pt-DNA adduct levels and chemoresistance in non-small cell lung (NSCLC) and breast cancer cell lines, and determined the same resistant mechanisms, such as cell uptake/efflux, intracellular inactivation and DNA repair. Carboplatin had partially different resistant mechanisms. CONCLUSION: Functional analysis of major resistant steps can identify some chemoresistance mechanisms that can potentially help design personalized chemotherapy to overcome resistance. This approach can be applied to several different cancer types. A Phase 0 microdosing clinical trial is currently going on in patients with NSCLC and bladder cancer. Citation Format: Amy W. Pan, Sisi Wang, Hongyong Zhang, Ruth Vinall, Tzu-yin Lin, Michael Malfatti, Maike Zimmermann, Tiffany Scharadin, Kenneth Turteltaub, Ralph de Vere White, Chong-xian Pan, Paul Henderson. Molecular dissection of platinum resistance through functional analysis. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 905. doi:10.1158/1538-7445.AM2014-905

Collaboration


Dive into the Maike Zimmermann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chong Xian Pan

University of California

View shared research outputs
Top Co-Authors

Avatar

Hongyong Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar

Kenneth W. Turteltaub

Lawrence Livermore National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Michael A. Malfatti

Lawrence Livermore National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary C. Horne

University of California

View shared research outputs
Top Co-Authors

Avatar

Sisi Wang

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge