Network


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

Hotspot


Dive into the research topics where Martin Thunemann is active.

Publication


Featured researches published by Martin Thunemann.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Anemia and splenomegaly in cGKI-deficient mice

Michael Föller; Susanne Feil; Kamran Ghoreschi; Saisudha Koka; Andrea Gerling; Martin Thunemann; Franz Hofmann; Beat Schuler; Johannes Vogel; Bernd J. Pichler; Ravi S. Kasinathan; Jan P. Nicolay; Stephan M. Huber; Florian Lang; Robert Feil

To explore the functional significance of cGMP-dependent protein kinase type I (cGKI) in the regulation of erythrocyte survival, gene-targeted mice lacking cGKI were compared with their control littermates. By the age of 10 weeks, cGKI-deficient mice exhibited pronounced anemia and splenomegaly. Compared with control mice, the cGKI mutants had significantly lower red blood cell count, packed cell volume, and hemoglobin concentration. Anemia was associated with a higher reticulocyte number and an increase of plasma erythropoietin concentration. The spleens of cGKI mutant mice were massively enlarged and contained a higher fraction of Ter119+ erythroid cells, whereas the relative proportion of leukocyte subpopulations was not changed. The Ter119+ cGKI-deficient splenocytes showed a marked increase in annexin V binding, pointing to phosphatidylserine (PS) exposure at the outer membrane leaflet, a hallmark of suicidal erythrocyte death or eryptosis. Compared with control erythrocytes, cGKI-deficient erythrocytes exhibited in vitro a higher cytosolic Ca2+ concentration, a known trigger of eryptosis, and showed increased PS exposure, which was paralleled by a faster clearance in vivo. Together, these results identify a role of cGKI as mediator of erythrocyte survival and extend the emerging concept that cGMP/cGKI signaling has an antiapoptotic/prosurvival function in a number of cell types in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Intercellular signaling via cyclic GMP diffusion through gap junctions restarts meiosis in mouse ovarian follicles.

Leia C. Shuhaibar; Jeremy R. Egbert; Rachael P. Norris; Paul D. Lampe; Viacheslav O. Nikolaev; Martin Thunemann; Lai Wen; Robert Feil; Laurinda A. Jaffe

Significance By imaging cyclic GMP (cGMP) in live ovarian follicles from mice, we show how luteinizing hormone signaling in the follicle periphery results in a rapid decrease in cGMP in the oocyte, thus reinitiating meiosis. Luteinizing hormone signaling lowers cGMP in the outer cells of the follicle, then cGMP in the oocyte decreases as a consequence of diffusion through gap junctions. These findings demonstrate directly that a physiological signal initiated by a stimulus in one region of an intact tissue can travel across many layers of cells via cyclic nucleotide diffusion through gap junctions. Meiosis in mammalian oocytes is paused until luteinizing hormone (LH) activates receptors in the mural granulosa cells of the ovarian follicle. Prior work has established the central role of cyclic GMP (cGMP) from the granulosa cells in maintaining meiotic arrest, but it is not clear how binding of LH to receptors that are located up to 10 cell layers away from the oocyte lowers oocyte cGMP and restarts meiosis. Here, by visualizing intercellular trafficking of cGMP in real-time in live follicles from mice expressing a FRET sensor, we show that diffusion of cGMP through gap junctions is responsible not only for maintaining meiotic arrest, but also for rapid transmission of the signal that reinitiates meiosis from the follicle surface to the oocyte. Before LH exposure, the cGMP concentration throughout the follicle is at a uniformly high level of ∼2–4 μM. Then, within 1 min of LH application, cGMP begins to decrease in the peripheral granulosa cells. As a consequence, cGMP from the oocyte diffuses into the sink provided by the large granulosa cell volume, such that by 20 min the cGMP concentration in the follicle is uniformly low, ∼100 nM. The decrease in cGMP in the oocyte relieves the inhibition of the meiotic cell cycle. This direct demonstration that a physiological signal initiated by a stimulus in one region of an intact tissue can travel across many layers of cells via cyclic nucleotide diffusion through gap junctions could provide a general mechanism for diverse cellular processes.


Circulation Research | 2013

Transgenic mice for cGMP imaging.

Martin Thunemann; Lai Wen; Matthias Hillenbrand; Susanne Feil; Thomas Ott; Xiaoxing Han; Dai Fukumura; Rakesh K. Jain; Michael Russwurm; Cor de Wit; Robert Feil

Rationale: Cyclic GMP (cGMP) is an important intracellular signaling molecule in the cardiovascular system, but its spatiotemporal dynamics in vivo is largely unknown. Objective: To generate and characterize transgenic mice expressing the fluorescence resonance energy transfer–based ratiometric cGMP sensor, cGMP indicator with an EC50 of 500 nmol/L (cGi500), in cardiovascular tissues. Methods and Results: Mouse lines with smooth muscle–specific or ubiquitous expression of cGi500 were generated by random transgenesis using an SM22&agr; promoter fragment or by targeted integration of a Cre recombinase–activatable expression cassette driven by the cytomegalovirus early enhancer/chicken &bgr;-actin/&bgr;-globin promoter into the Rosa26 locus, respectively. Primary smooth muscle cells isolated from aorta, bladder, and colon of cGi500 mice showed strong sensor fluorescence. Basal cGMP concentrations were <100 nmol/L, whereas stimulation with cGMP-elevating agents such as 2-(N,N-diethylamino)-diazenolate-2-oxide diethylammonium salt (DEA/NO) or the natriuretic peptides, atrial natriuretic peptide, and C-type natriuretic peptide evoked fluorescence resonance energy transfer changes corresponding to cGMP peak concentrations of ≈3 µmol/L. However, different types of smooth muscle cells had different sensitivities of their cGMP responses to DEA/NO, atrial natriuretic peptide, and C-type natriuretic peptide. Robust nitric oxide–induced cGMP transients with peak concentrations of ≈1 to >3 µmol/L could also be monitored in blood vessels of the isolated retina and in the cremaster microcirculation of anesthetized mice. Moreover, with the use of a dorsal skinfold chamber model and multiphoton fluorescence resonance energy transfer microscopy, nitric oxide–stimulated vascular cGMP signals associated with vasodilation were detected in vivo in an acutely untouched preparation. Conclusions: These cGi500 transgenic mice permit the visualization of cardiovascular cGMP signals in live cells, tissues, and mice under normal and pathological conditions or during pharmacotherapy with cGMP-elevating drugs.


Nature Neuroscience | 2017

Microglia turnover with aging and in an Alzheimer's model via long-term in vivo single-cell imaging

Petra Füger; Jasmin K. Hefendehl; Karthik Veeraraghavalu; Ann-Christin Wendeln; Christine Schlosser; Ulrike Obermüller; Bettina M. Wegenast-Braun; Jonas J. Neher; Peter Martus; Shinichi Kohsaka; Martin Thunemann; Robert Feil; Sangram S. Sisodia; Angelos Skodras; Mathias Jucker

To clarify the role of microglia in brain homeostasis and disease, an understanding of their maintenance, proliferation and turnover is essential. The lifespan of brain microglia, however, remains uncertain, and reflects confounding factors in earlier assessments that were largely indirect. We genetically labeled single resident microglia in living mice and then used multiphoton microscopy to monitor these cells over time. Under homeostatic conditions, we found that neocortical resident microglia were long-lived, with a median lifetime of well over 15 months; thus, approximately half of these cells survive the entire mouse lifespan. While proliferation of resident neocortical microglia under homeostatic conditions was low, microglial proliferation in a mouse model of Alzheimers β-amyloidosis was increased threefold. The persistence of individual microglia throughout the mouse lifespan provides an explanation for how microglial priming early in life can induce lasting functional changes and how microglial senescence may contribute to age-related neurodegenerative diseases.


eLife | 2016

Cell type specificity of neurovascular coupling in cerebral cortex

Hana Uhlirova; Kıvılcım Kılıç; Peifang Tian; Martin Thunemann; Michèle Desjardins; Payam A. Saisan; Sava Sakadžić; Torbjørn V. Ness; Celine Mateo; Qun Cheng; Kimberly L. Weldy; Florence Razoux; Matthieu Vandenberghe; Jonathan A. Cremonesi; Christopher G. L. Ferri; Krystal Nizar; Vishnu B. Sridhar; Tyler Steed; Maxim Abashin; Yeshaiahu Fainman; Eliezer Masliah; Srdjan Djurovic; Ole A. Andreassen; Gabriel A. Silva; David A. Boas; David Kleinfeld; Richard B. Buxton; Gaute T. Einevoll; Anders M. Dale; Anna Devor

Identification of the cellular players and molecular messengers that communicate neuronal activity to the vasculature driving cerebral hemodynamics is important for (1) the basic understanding of cerebrovascular regulation and (2) interpretation of functional Magnetic Resonance Imaging (fMRI) signals. Using a combination of optogenetic stimulation and 2-photon imaging in mice, we demonstrate that selective activation of cortical excitation and inhibition elicits distinct vascular responses and identify the vasoconstrictive mechanism as Neuropeptide Y (NPY) acting on Y1 receptors. The latter implies that task-related negative Blood Oxygenation Level Dependent (BOLD) fMRI signals in the cerebral cortex under normal physiological conditions may be mainly driven by the NPY-positive inhibitory neurons. Further, the NPY-Y1 pathway may offer a potential therapeutic target in cerebrovascular disease. DOI: http://dx.doi.org/10.7554/eLife.14315.001


Cell Reports | 2016

Sildenafil Potentiates a cGMP-Dependent Pathway to Promote Melanoma Growth

Sandeep Dhayade; Susanne Kaesler; Tobias Sinnberg; Hyazinth Dobrowinski; Stefanie Peters; Ulrike Naumann; He Liu; Robert E. Hunger; Martin Thunemann; Tilo Biedermann; Birgit Schittek; Hans-Uwe Simon; Susanne Feil; Robert Feil

Sildenafil, an inhibitor of the cGMP-degrading phosphodiesterase 5 that is used to treat erectile dysfunction, has been linked to an increased risk of melanoma. Here, we have examined the potential connection between cGMP-dependent signaling cascades and melanoma growth. Using a combination of biochemical assays and real-time monitoring of melanoma cells, we report a cGMP-dependent growth-promoting pathway in murine and human melanoma cells. We document that C-type natriuretic peptide (CNP), a ligand of the membrane-bound guanylate cyclase B, enhances the activity of cGMP-dependent protein kinase I (cGKI) in melanoma cells by increasing the intracellular levels of cGMP. Activation of this cGMP pathway promotes melanoma cell growth and migration in a p44/42 MAPK-dependent manner. Sildenafil treatment further increases intracellular cGMP concentrations, potentiating activation of this pathway. Collectively, our data identify this cGMP-cGKI pathway as the link between sildenafil usage and increased melanoma risk.


Free Radical Biology and Medicine | 2012

H2O2 lowers the cytosolic Ca2+ concentration via activation of cGMP-dependent protein kinase Iα

Paul Markus Müller; Robert Gnügge; Sandeep Dhayade; Martin Thunemann; Peter Krippeit-Drews; Gisela Drews; Robert Feil

The cGMP-dependent protein kinase I (cGKI) is a key mediator of cGMP signaling, but the specific functions of its two isoforms, cGKIα and cGKIβ, are poorly understood. Recent studies indicated a novel cGMP-independent role for cGKIα in redox sensing. To dissect the effects of oxidative stress on the cGKI isoforms, we used mouse embryonic fibroblasts and vascular smooth muscle cells (VSMCs) expressing both, one, or none of them. In cGKIα-expressing cells, but not in cells expressing only cGKIβ, incubation with H₂O₂ induced the formation of a disulfide bond between the two identical subunits of the dimeric enzyme. Oxidation of cGKIα was associated with increased phosphorylation of its substrate, vasodilator-stimulated phosphoprotein. H₂O₂ did not stimulate cGMP production, indicating that it activates cGKIα directly via oxidation. Interestingly, there was a mutual influence of H₂O₂ and cGMP on cGKI activity and disulfide bond formation, respectively; preoxidation of the kinase with H₂O₂ slightly impaired its activation by cGMP, whereas preactivation of the enzyme with cGMP attenuated its oxidation by H₂O₂. To evaluate the functional relevance of the noncanonical H₂O₂-cGKIα pathway, we studied the regulation of the cytosolic Ca²⁺ concentration ([Ca²⁺](i)). H₂O₂ suppressed norepinephrine-induced Ca²⁺ transients in cGKIα-expressing VSMCs and, to a lower extent, in VSMCs expressing only cGKIβ or none of the isoforms. Thus, H₂O₂ lowers [Ca²⁺](i) mainly via a cGKIα-dependent pathway. These results indicate that oxidative stress selectively targets the cGKIα isoform, which then modulates cellular processes in a cGMP-independent manner. A decrease in [Ca²⁺](i) in VSMCs via activation of cGKIα might be a major mechanism of H₂O₂-induced vasodilation.


Philosophical Transactions of the Royal Society B | 2016

The roadmap for estimation of cell-type-specific neuronal activity from non-invasive measurements

Hana Uhlirova; Kıvılcım Kılıç; Peifang Tian; Sava Sakadžić; Louis Gagnon; Martin Thunemann; Michèle Desjardins; Payam A. Saisan; Krystal Nizar; Mohammad A. Yaseen; Donald J. Hagler; Matthieu Vandenberghe; Srdjan Djurovic; Ole A. Andreassen; Gabriel A. Silva; Eliezer Masliah; David Kleinfeld; Sergei A. Vinogradov; Richard B. Buxton; Gaute T. Einevoll; David A. Boas; Anders M. Dale; Anna Devor

The computational properties of the human brain arise from an intricate interplay between billions of neurons connected in complex networks. However, our ability to study these networks in healthy human brain is limited by the necessity to use non-invasive technologies. This is in contrast to animal models where a rich, detailed view of cellular-level brain function with cell-type-specific molecular identity has become available due to recent advances in microscopic optical imaging and genetics. Thus, a central challenge facing neuroscience today is leveraging these mechanistic insights from animal studies to accurately draw physiological inferences from non-invasive signals in humans. On the essential path towards this goal is the development of a detailed ‘bottom-up’ forward model bridging neuronal activity at the level of cell-type-specific populations to non-invasive imaging signals. The general idea is that specific neuronal cell types have identifiable signatures in the way they drive changes in cerebral blood flow, cerebral metabolic rate of O2 (measurable with quantitative functional Magnetic Resonance Imaging), and electrical currents/potentials (measurable with magneto/electroencephalography). This forward model would then provide the ‘ground truth’ for the development of new tools for tackling the inverse problem—estimation of neuronal activity from multimodal non-invasive imaging data. This article is part of the themed issue ‘Interpreting BOLD: a dialogue between cognitive and cellular neuroscience’.


Frontiers in Physiology | 2014

Correlative intravital imaging of cGMP signals and vasodilation in mice

Martin Thunemann; Kjestine Schmidt; Cor de Wit; Xiaoxing Han; Rakesh K. Jain; Dai Fukumura; Robert Feil

Cyclic guanosine monophosphate (cGMP) is an important signaling molecule and drug target in the cardiovascular system. It is well known that stimulation of the vascular nitric oxide (NO)-cGMP pathway results in vasodilation. However, the spatiotemporal dynamics of cGMP signals themselves and the cGMP concentrations within specific cardiovascular cell types in health, disease, and during pharmacotherapy with cGMP-elevating drugs are largely unknown. To facilitate the analysis of cGMP signaling in vivo, we have generated transgenic mice that express fluorescence resonance energy transfer (FRET)-based cGMP sensor proteins. Here, we describe two models of intravital FRET/cGMP imaging in the vasculature of cGMP sensor mice: (1) epifluorescence-based ratio imaging in resistance-type vessels of the cremaster muscle and (2) ratio imaging by multiphoton microscopy within the walls of subcutaneous blood vessels accessed through a dorsal skinfold chamber. Both methods allow simultaneous monitoring of NO-induced cGMP transients and vasodilation in living mice. Detailed protocols of all steps necessary to perform and evaluate intravital imaging experiments of the vasculature of anesthetized mice including surgery, imaging, and data evaluation are provided. An image segmentation approach is described to estimate FRET/cGMP changes within moving structures such as the vessel wall during vasodilation. The methods presented herein should be useful to visualize cGMP or other biochemical signals that are detectable with FRET-based biosensors, such as cyclic adenosine monophosphate or Ca2+, and to correlate them with respective vascular responses. With further refinement and combination of transgenic mouse models and intravital imaging technologies, we envision an exciting future, in which we are able to “watch” biochemistry, (patho-)physiology, and pharmacotherapy in the context of a living mammalian organism.


Methods of Molecular Biology | 2014

Genetic inducible fate mapping in adult mice using tamoxifen-dependent Cre recombinases.

Susanne Feil; Jana Krauss; Martin Thunemann; Robert Feil

The Cre/lox site-specific recombination system allows the control of gene activity in space and time in almost any tissue of the mouse. A major technical advance was the development of tamoxifen-dependent Cre recombinases, such as CreER(T2), that can be activated by administration of tamoxifen to the animal. This powerful tool greatly facilitates the study of gene functions and the generation of more realistic animal models of sporadic human diseases. Another important application of tamoxifen-dependent Cre recombinases is genetic inducible fate mapping (GIFM). In GIFM studies, the inducible Cre/lox system is used to genetically label a defined cell population at a selected time by irreversible activation of the expression of a Cre-responsive reporter transgene. Then, marked cells are detected at later time points to determine how the originally labeled progenitors contribute to specific structures and cell types during pre- and postnatal development. GIFM was initially applied during mouse embryogenesis, but is now increasingly used for cell lineage tracing in adult mice under physiological and pathophysiological conditions. Here we describe the design of GIFM experiments in adult mice as exemplified by CreER(T2)-assisted tracing of vascular smooth muscle cells during the development of atherosclerotic lesions. First, we give an overview of reporter transgenes available for genetic cell marking that are expressed from the Rosa26 locus, such as β-galactosidase and fluorescent proteins. Then we present detailed protocols for the generation of experimental mice for GIFM studies, the induction of cell labeling by tamoxifen treatment, and the detection of marked cells in fixed and live tissues. Each section also provides a discussion of limitations and common pitfalls of GIFM experiments. Most of the protocols can be easily adapted to other developmental stages, cell types, Cre recombinases, and reporter transgenes and, thus, can be used as general guidelines for GIFM studies in mice.

Collaboration


Dive into the Martin Thunemann's collaboration.

Top Co-Authors

Avatar

Robert Feil

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Anna Devor

University of California

View shared research outputs
Top Co-Authors

Avatar

Susanne Feil

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Anders M. Dale

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lai Wen

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hana Uhlirova

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge