Network


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

Hotspot


Dive into the research topics where Anna-Kristin Ludwig is active.

Publication


Featured researches published by Anna-Kristin Ludwig.


Colloids and Surfaces B: Biointerfaces | 2011

Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy.

Viktoriya Sokolova; Anna-Kristin Ludwig; Sandra Hornung; Olga Rotan; Peter A. Horn; Matthias Epple; Bernd Giebel

Exosomes from three different cell types (HEK 293T, ECFC, MSC) were characterised by scanning electron microscopy (SEM), dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA). The diameter was around 110 nm for the three cell types. The stability of exosomes was examined during storage at -20°C, 4°C, and 37°C. The size of the exosomes decreased at 4°C and 37°C, indicating a structural change or degradation. Multiple freezing to -20°C and thawing did not affect the exosome size. Multiple ultracentrifugation also did not change the exosome size.


Leukemia | 2014

MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease

Lambros Kordelas; Rebmann; Anna-Kristin Ludwig; Radtke S; Johannes Ruesing; Doeppner Tr; Matthias Epple; Peter A. Horn; Dietrich W. Beelen; Bernd Giebel

GV, FF and AI designed the research and wrote the manuscript; FDR, MRC, CR and GS treated the patients, collected the data and commented on the manuscript; FL and AV collected and analyzed the data and commented on the manuscript; PPP and MR performed the statistical analysis. AG, MR, and MAL generated GEP; FF performed GEP data analysis; SP performed EBM diagnostic accuracy analysis; PPP, SAP, AI and GV designed the molecular analysis and funded it, analyzed GEP data and wrote the manuscript.


The International Journal of Biochemistry & Cell Biology | 2012

Exosomes: Small vesicles participating in intercellular communication

Anna-Kristin Ludwig; Bernd Giebel

Exosomes are small membrane vesicles, which eukaryotic cells secrete into their extracellular environment. They are formed as intraluminal vesicles by inward budding of the limiting membrane into the lumen of late endosomes. Upon fusion of thus arising multivesicular bodies with the plasma membrane, these vesicles are released as exosomes and enter body fluids such as blood plasma, urine and saliva. Containing certain combinations of lipids, adhesion and intercellular signaling molecules as well as RNAs, exosomes participate in intercellular communication processes. Depending on their origin, exosomes can modulate immune-regulatory processes, set up tumor escape mechanisms and mediate regenerative or degenerative processes, amongst others. In summary, exosomes are molecular complex intercellular signaling organelles with multiple functions, which appear as promising new tools for the clinical diagnostics and potentially for novel therapeutic strategies.


Stem Cells Translational Medicine | 2015

Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression

Thorsten R. Doeppner; Josephine Herz; André Görgens; Jana Schlechter; Anna-Kristin Ludwig; Stefan Radtke; Kyra de Miroschedji; Peter A. Horn; Bernd Giebel; Dirk M. Hermann

Although the initial concepts of stem cell therapy aimed at replacing lost tissue, more recent evidence has suggested that stem and progenitor cells alike promote postischemic neurological recovery by secreted factors that restore the injured brains capacity to reshape. Specifically, extracellular vesicles (EVs) derived from stem cells such as exosomes have recently been suggested to mediate restorative stem cell effects. In order to define whether EVs indeed improve postischemic neurological impairment and brain remodeling, we systematically compared the effects of mesenchymal stem cell (MSC)‐derived EVs (MSC‐EVs) with MSCs that were i.v. delivered to mice on days 1, 3, and 5 (MSC‐EVs) or on day 1 (MSCs) after focal cerebral ischemia in C57BL6 mice. For as long as 28 days after stroke, motor coordination deficits, histological brain injury, immune responses in the peripheral blood and brain, and cerebral angiogenesis and neurogenesis were analyzed. Improved neurological impairment and long‐term neuroprotection associated with enhanced angioneurogenesis were noticed in stroke mice receiving EVs from two different bone marrow‐derived MSC lineages. MSC‐EV administration closely resembled responses to MSCs and persisted throughout the observation period. Although cerebral immune cell infiltration was not affected by MSC‐EVs, postischemic immunosuppression (i.e., B‐cell, natural killer cell, and T‐cell lymphopenia) was attenuated in the peripheral blood at 6 days after ischemia, providing an appropriate external milieu for successful brain remodeling. Because MSC‐EVs have recently been shown to be apparently safe in humans, the present study provides clinically relevant evidence warranting rapid proof‐of‐concept studies in stroke patients.


Stem Cells and Development | 2010

Interferon-γ and Tumor Necrosis Factor-α Differentially Affect Cytokine Expression and Migration Properties of Mesenchymal Stem Cells

Hatim Hemeda; Mark Jakob; Anna-Kristin Ludwig; Bernd Giebel; Stephan Lang; Sven Brandau

Mesenchymal stem cells (MSCs) are multipotent progenitor cells with the capacity to differentiate into different tissue cell types such as chondrocytes, osteocytes, and adipocytes. In addition, they can home to damaged, in-flamed, and malignant tissues and display immunomodulatory properties. Since tissue-derived factors might modulate these properties, we decided to explore the impact of prototypic tissue-derived inflammatory cytokines such as TNF-alpha and IFN-gamma on immunomodulatory MSCs functions. To this end, we used primary bone marrow and cord blood-derived MSCs as well as an immortalized MSC line (V54/2) as model systems. We demonstrate that under unstimulated conditions, V54/2 cells constitutively express low levels of indoleamine 2,3-dioxygenase (IDO), exert an immunosuppressive effect on activated T-lymphocyte proliferation, secrete a distinct set of cytokines, and express a wide range of chemokine receptors. Upon stimulation, the proinflammatory cytokines IFN-gamma and TNF-alpha did not inhibit suppression of T-cell proliferation, although IDO expression was up-regulated by IFN-gamma. In contrast, TNF-alpha but not IFN-gamma amplified the cytokine production of V54/2 and primary MSCs. Interestingly, IFN-gamma was superior to TNF-alpha in up-regulating expression of chemokine receptors and migration of the V54/2 cell line, while TNF-alpha was the predominant regulator of migration in primary MSCs. Altogether, our data show that properties of MSCs depend on local environmental factors. In particular, we have shown that IFN-gamma and TNF-alpha differentially regulate cytokine expression and migration of MSCs.


Stem Cells | 2012

Transduction of Neural Precursor Cells with TAT‐Heat Shock Protein 70 Chaperone: Therapeutic Potential Against Ischemic Stroke after Intrastriatal and Systemic Transplantation

Thorsten R. Doeppner; Tobias A. S. Ewert; Lars Tönges; Josephine Herz; Anil Zechariah; Ayman ElAli; Anna-Kristin Ludwig; Bernd Giebel; Florian Nagel; Gunnar P.H. Dietz; Jens Weise; Dirk M. Hermann; Mathias Bähr

Novel therapeutic concepts against cerebral ischemia focus on cell‐based therapies in order to overcome some of the side effects of thrombolytic therapy. However, cell‐based therapies are hampered because of restricted understanding regarding optimal cell transplantation routes and due to low survival rates of grafted cells. We therefore transplanted adult green fluorescence protein positive neural precursor cells (NPCs) either intravenously (systemic) or intrastriatally (intracerebrally) 6 hours after stroke in mice. To enhance survival of NPCs, cells were in vitro protein‐transduced with TAT‐heat shock protein 70 (Hsp70) before transplantation followed by a systematic analysis of brain injury and underlying mechanisms depending on cell delivery routes. Transduction of NPCs with TAT‐Hsp70 resulted in increased intracerebral numbers of grafted NPCs after intracerebral but not after systemic transplantation. Whereas systemic delivery of either native or transduced NPCs yielded sustained neuroprotection and induced neurological recovery, only TAT‐Hsp70‐transduced NPCs prevented secondary neuronal degeneration after intracerebral delivery that was associated with enhanced functional outcome. Furthermore, intracerebral transplantation of TAT‐Hsp70‐transduced NPCs enhanced postischemic neurogenesis and induced sustained high levels of brain‐derived neurotrophic factor, glial cell line‐derived neurotrophic factor, and vascular endothelial growth factor in vivo. Neuroprotection after intracerebral cell delivery correlated with the amount of surviving NPCs. On the contrary, systemic delivery of NPCs mediated acute neuroprotection via stabilization of the blood‐brain‐barrier, concomitant with reduced activation of matrix metalloprotease 9 and decreased formation of reactive oxygen species. Our findings imply two different mechanisms of action of intracerebrally and systemically transplanted NPCs, indicating that systemic NPC delivery might be more feasible for translational stroke concepts, lacking a need of in vitro manipulation of NPCs to induce long‐term neuroprotection. STEM CELLS2012;30:1297–1310


Stem Cells Translational Medicine | 2016

Mesenchymal Stromal Cell-Derived Extracellular Vesicles Protect the Fetal Brain After Hypoxia-Ischemia

Daan R. M. G. Ophelders; Tim G. A. M. Wolfs; Reint K. Jellema; Alex Zwanenburg; Peter Andriessen; Tammo Delhaas; Anna-Kristin Ludwig; Stefan Radtke; Vera Peters; Leon Janssen; Bernd Giebel; Boris W. Kramer

Preterm neonates are susceptible to perinatal hypoxic‐ischemic brain injury, for which no treatment is available. In a preclinical animal model of hypoxic‐ischemic brain injury in ovine fetuses, we have demonstrated the neuroprotective potential of systemically administered mesenchymal stromal cells (MSCs). The mechanism of MSC treatment is unclear but suggested to be paracrine, through secretion of extracellular vesicles (EVs). Therefore, we investigated in this study the protective effects of mesenchymal stromal cell‐derived extracellular vesicles (MSC‐EVs) in a preclinical model of preterm hypoxic‐ischemic brain injury. Ovine fetuses were subjected to global hypoxia‐ischemia by transient umbilical cord occlusion, followed by in utero intravenous administration of MSC‐EVs. The therapeutic effects of MSC‐EV administration were assessed by analysis of electrophysiological parameters and histology of the brain. Systemic administration of MSC‐EVs improved brain function by reducing the total number and duration of seizures, and by preserving baroreceptor reflex sensitivity. These functional protections were accompanied by a tendency to prevent hypomyelination. Cerebral inflammation remained unaffected by the MSC‐EV treatment. Our data demonstrate that MSC‐EV treatment might provide a novel strategy to reduce the neurological sequelae following hypoxic‐ischemic injury of the preterm brain. Our study results suggest that a cell‐free preparation comprising neuroprotective MSC‐EVs could substitute MSCs in the treatment of preterm neonates with hypoxic‐ischemic brain injury, thereby circumventing the potential risks of systemic administration of living cells.


Brain Behavior and Immunity | 2017

Mesenchymal stem cell-derived extracellular vesicles ameliorate inflammation-induced preterm brain injury

Karla Drommelschmidt; Meray Serdar; Ivo Bendix; Josephine Herz; Frederik Bertling; Sebastian Prager; Matthias Keller; Anna-Kristin Ludwig; Vikas Duhan; Stefan Radtke; Kyra de Miroschedji; Peter A. Horn; Yohan van de Looij; Bernd Giebel; Ursula Felderhoff-Müser

OBJECTIVE Preterm brain injury is a major cause of disability in later life, and may result in motor, cognitive and behavioural impairment for which no treatment is currently available. The aetiology is considered as multifactorial, and one underlying key player is inflammation leading to white and grey matter injury. Extracellular vesicles secreted by mesenchymal stem/stromal cells (MSC-EVs) have shown therapeutic potential in regenerative medicine. Here, we investigated the effects of MSC-EV treatment on brain microstructure and maturation, inflammatory processes and long-time outcome in a rodent model of inflammation-induced brain injury. METHODS 3-Day-old Wistar rats (P3) were intraperitoneally injected with 0.25mg/kg lipopolysaccharide or saline and treated with two repetitive doses of 1×108 cell equivalents of MSC-EVs per kg bodyweight. Cellular degeneration and reactive gliosis at P5 and myelination at P11 were evaluated by immunohistochemistry and western blot. Long-term cognitive and motor function was assessed by behavioural testing. Diffusion tensor imaging at P125 evaluated long-term microstructural white matter alterations. RESULTS MSC-EV treatment significantly ameliorated inflammation-induced neuronal cellular degeneration reduced microgliosis and prevented reactive astrogliosis. Short-term myelination deficits and long-term microstructural abnormalities of the white matter were restored by MSC-EV administration. Morphological effects of MSC-EV treatment resulted in improved long-lasting cognitive functions INTERPRETATION: MSC-EVs ameliorate inflammation-induced cellular damage in a rat model of preterm brain injury. MSC-EVs may serve as a novel therapeutic option by prevention of neuronal cell death, restoration of white matter microstructure, reduction of gliosis and long-term functional improvement.


The International Journal of Biochemistry & Cell Biology | 2012

Lipid raft redistribution and morphological cell polarization are separable processes providing a basis for hematopoietic stem and progenitor cell migration

André Görgens; Julia Beckmann; Anna-Kristin Ludwig; Michael Möllmann; Jan Dürig; Peter A. Horn; Lawrence Rajendran; Bernd Giebel

Freshly isolated human hematopoietic stem and progenitor cells (HSPCs) are small and round cells which upon cultivation adopt a polarized morphology and redistribute certain cell surface antigens. To functionally dissect this polarization process, we addressed impacts of protein synthesis, HSPC trafficking, cytoskeleton organization or lipid raft integrity on the establishment and maintenance of the cell polarity of human HSPCs. Effects on the morphology, sub-cellular distribution of lipid raft-associated molecular polarization markers (Flotillin-1, Flotillin-2, ICAM-3) and in vitro migration capabilities of treated cells were studied. We could distinguish two levels of cellular polarization, a molecular and a morphological level. Our data suggest that protein synthesis, lipid raft integrity and enzymatic activities of PI3K and aPKC are required to organize the molecular cell polarity. The morphological cell polarization process, however, also depends on actin polymerization and rho-GTPase activities. In summary, our data qualify HSPC polarization processes as new pharmaceutical target to interfere with migratory and with homing capabilities of HSPCs.


Archive | 2013

Use of Preparations Comprising Exosomes Derived From Mesenchymal Stem Cells (MSCs) in the Prevention and Therapy of Inflammatory Conditions

Dietrich Wilhelm Beelen; Thorsten Deoppner; Ursula Felderhoff-Mueser; Bernd Giebel; Dirk M. Hermann; Peter A. Horn; Matthias Keller; Lambros Kordelas; Anna-Kristin Ludwig; Vera Rebmann

Collaboration


Dive into the Anna-Kristin Ludwig's collaboration.

Top Co-Authors

Avatar

Bernd Giebel

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

André Görgens

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Dirk M. Hermann

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Josephine Herz

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Jan Dürig

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Kyra de Miroschedji

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Lambros Kordelas

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Matthias Epple

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Michael Möllmann

University of Duisburg-Essen

View shared research outputs
Researchain Logo
Decentralizing Knowledge