Network


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

Hotspot


Dive into the research topics where Barbara Ludwig is active.

Publication


Featured researches published by Barbara Ludwig.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Transplantation of human islets without immunosuppression

Barbara Ludwig; Andreas Reichel; Anja Steffen; Baruch Zimerman; Andrew V. Schally; Norman L. Block; Clark K. Colton; Stefan Ludwig; Stephan Kersting; Ezio Bonifacio; Michele Solimena; Zohar Gendler; Avi Rotem; Uriel Barkai; Stefan R. Bornstein

Significance Diabetes mellitus type 1 is an autoimmune disease that results in irreversible destruction of insulin-producing beta cells. Substantial advances have been made in beta cell replacement therapies over the last decades. However, lack of eligible donor organs and the need for chronic immunosuppression to prevent rejection critically limit a widespread application of these strategies. In this paper we present the clinical success of using a bioartificial pancreas for the transplantation of insulin-producing islets without affecting the immune system. In a patient with long-standing type-1 diabetes we could demonstrate persistent graft function and regulated insulin secretion without the need for immune-modulating medication. This strategy opens up avenues for more widespread and safe application of various cell-based therapies. Transplantation of pancreatic islets is emerging as a successful treatment for type-1 diabetes. Its current stringent restriction to patients with critical metabolic lability is justified by the long-term need for immunosuppression and a persistent shortage of donor organs. We developed an oxygenated chamber system composed of immune-isolating alginate and polymembrane covers that allows for survival and function of islets without immunosuppression. A patient with type-1 diabetes received a transplanted chamber and was followed for 10 mo. Persistent graft function in this chamber system was demonstrated, with regulated insulin secretion and preservation of islet morphology and function without any immunosuppressive therapy. This approach may allow for future widespread application of cell-based therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Improvement of islet function in a bioartificial pancreas by enhanced oxygen supply and growth hormone releasing hormone agonist

Barbara Ludwig; Avi Rotem; Janine Schmid; Gordon C. Weir; Clark K. Colton; Mathias D. Brendel; Tova Neufeld; Norman L. Block; Karina Yavriyants; Anja Steffen; Stefan Ludwig; Triantafyllos Chavakis; Andreas Reichel; Dimitri Azarov; Baruch Zimermann; Shiri Maimon; Mariya Balyura; Tania Rozenshtein; Noa Shabtay; Pnina Vardi; Konstantin Bloch; Paul de Vos; Andrew V. Schally; Stefan R. Bornstein; Uriel Barkai

Islet transplantation is a feasible therapeutic alternative for metabolically labile patients with type 1 diabetes. The primary therapeutic target is stable glycemic control and prevention of complications associated with diabetes by reconstitution of endogenous insulin secretion. However, critical shortage of donor organs, gradual loss in graft function over time, and chronic need for immunosuppression limit the indication for islet transplantation to a small group of patients. Here we present a promising approach to address these limitations by utilization of a macrochamber specially engineered for islet transplantation. The s.c. implantable device allows for controlled and adequate oxygen supply and provides immunological protection of donor islets against the host immune system. The minimally invasive implantable chamber normalized blood glucose in streptozotocin-induced diabetic rodents for up to 3 mo. Sufficient graft function depended on oxygen supply. Pretreatment with the growth hormone-releasing hormone (GHRH) agonist, JI-36, significantly enhanced graft function by improving glucose tolerance and increasing β-cell insulin reserve in rats thereby allowing for a reduction of the islet mass required for metabolic control. As a result of hypervascularization of the tissue surrounding the device, no relevant delay in insulin response to glucose changes has been observed. Consequently, this system opens up a fundamental strategy for therapy of diabetes and may provide a promising avenue for future approaches to xenotransplantation.


Cell Transplantation | 2013

Enhanced Oxygen Supply Improves Islet Viability in a New Bioartificial Pancreas

Uriel Barkai; Gordon C. Weir; Clark K. Colton; Barbara Ludwig; Stefan R. Bornstein; Mathias D. Brendel; Tova Neufeld; Chezi Bremer; Assaf Leon; Yoav Evron; Karina Yavriyants; Dimitri Azarov; Baruch Zimermann; Shiri Maimon; Noa Shabtay; Maria Balyura; Tania Rozenshtein; Pnina Vardi; Konstantin Bloch; Paul de Vos; Avi Rotem

The current epidemic of diabetes with its overwhelming burden on our healthcare system requires better therapeutic strategies. Here we present a promising novel approach for a curative strategy that may be accessible for all insulin-dependent diabetes patients. We designed a subcutaneous implantable bioartificial pancreas (BAP)—the “β-Air”—that is able to overcome critical challenges in current clinical islet transplantation protocols: adequate oxygen supply to the graft and protection of donor islets against the host immune system. The system consists of islets of Langerhans immobilized in an alginate hydrogel, a gas chamber, a gas permeable membrane, an external membrane, and a mechanical support. The minimally invasive implantable device, refueled with oxygen via subdermally implanted access ports, completely normalized diabetic indicators of glycemic control (blood glucose intravenous glucose tolerance test and HbA1c) in streptozotocin-induced diabetic rats for periods up to 6 months. The functionality of the device was dependent on oxygen supply to the device as the grafts failed when oxygen supply was ceased. In addition, we showed that the device is immunoprotective as it allowed for survival of not only isografts but also of allografts. Histological examination of the explanted devices demonstrated morphologically and functionally intact islets; the surrounding tissue was without signs of inflammation and showed visual evidence of vasculature at the site of implantation. Further increase in islets loading density will justify the translation of the system to clinical trials, opening up the potential for a novel approach in diabetes therapy.


PLOS ONE | 2013

The Efficacy of an Immunoisolating Membrane System for Islet Xenotransplantation in Minipigs

Tova Neufeld; Barbara Ludwig; Uriel Barkai; Gordon C. Weir; Clark K. Colton; Yoav Evron; Maria Balyura; Karina Yavriyants; Baruch Zimermann; Dmitri Azarov; Shiri Maimon; Noa Shabtay; Tania Rozenshtein; Dana Lorber; Anja Steffen; Udi Willenz; Konstantine Bloch; Pnina Vardi; Ran Taube; Paul de Vos; Eli C. Lewis; Stefan R. Bornstein; Avi Rotem

Developing a device that protects xenogeneic islets to allow treatment and potentially cure of diabetes in large mammals has been a major challenge in the past decade. Using xenogeneic islets for transplantation is required in light of donor shortage and the large number of diabetic patients that qualify for islet transplantation. Until now, however, host immunoreactivity against the xenogeneic graft has been a major drawback for the use of porcine islets. Our study demonstrates the applicability of a novel immunoprotective membrane that allows successful xenotransplantation of rat islets in diabetic minipigs without immunosuppressive therapy. Rat pancreatic islets were encapsulated in highly purified alginate and integrated into a plastic macrochamber covered by a poly-membrane for subcutaneous transplantation. Diabetic Sinclair pigs were transplanted and followed for up to 90 days. We demonstrated a persistent graft function and restoration of normoglycemia without the need for immunosuppressive therapy. This concept could potentially offer an attractive strategy for a more widespread islet replacement therapy that would restore endogenous insulin secretion in diabetic patients without the need for immunosuppressive drugs and may even open up an avenue for safe utilization of xenogeneic islet donors.


Journal of Immunology | 2013

The Complement Anaphylatoxin C5a Receptor Contributes to Obese Adipose Tissue Inflammation and Insulin Resistance

Julia Phieler; Kyoung-Jin Chung; Antonios Chatzigeorgiou; Anne Klotzsche-von Ameln; Ruben Garcia-Martin; David Sprott; Maria Moisidou; Theodora Tzanavari; Barbara Ludwig; Elena Baraban; Monika Ehrhart-Bornstein; Stefan R. Bornstein; Hassan Mziaut; Michele Solimena; Katia P. Karalis; Matina Economopoulou; John D. Lambris; Triantafyllos Chavakis

Obese adipose tissue (AT) inflammation contributes critically to development of insulin resistance. The complement anaphylatoxin C5a receptor (C5aR) has been implicated in inflammatory processes and as regulator of macrophage activation and polarization. However, the role of C5aR in obesity and AT inflammation has not been addressed. We engaged the model of diet-induced obesity and found that expression of C5aR was significantly upregulated in the obese AT, compared with lean AT. In addition, C5a was present in obese AT in the proximity of macrophage-rich crownlike structures. C5aR-sufficient and -deficient mice were fed a high-fat diet (HFD) or a normal diet (ND). C5aR deficiency was associated with increased AT weight upon ND feeding in males, but not in females, and with increased adipocyte size upon ND and HFD conditions in males. However, obese C5aR−/− mice displayed improved systemic and AT insulin sensitivity. Improved AT insulin sensitivity in C5aR−/− mice was associated with reduced accumulation of total and proinflammatory M1 macrophages in the obese AT, increased expression of IL-10, and decreased AT fibrosis. In contrast, no difference in β cell mass was observed owing to C5aR deficiency under an HFD. These results suggest that C5aR contributes to macrophage accumulation and M1 polarization in the obese AT and thereby to AT dysfunction and development of AT insulin resistance.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Agonist of growth hormone-releasing hormone as a potential effector for survival and proliferation of pancreatic islets

Barbara Ludwig; Christian G. Ziegler; Andrew V. Schally; Claudius Richter; Anja Steffen; Normund Jabs; Richard Funk; Mathias D. Brendel; Norman L. Block; Monika Ehrhart-Bornstein; Stefan R. Bornstein

Therapeutic strategies for transplantation of pancreatic islet cells are urgently needed to expand β-cell mass by stimulating islet cell proliferation and/or prolonging islet cell survival. Control of the islets by different growth factors provides a potential venue for augmenting β-cell mass. In the present study, we show the expression of the biologically active splice variant-1 (SV-1) of growth hormone-releasing hormone (GHRH) receptor in rat insulinoma (INS-1) cells as well as in rat and human pancreatic islets. In studies in vitro of INS-1 cells, the GHRH agonist JI-36 caused a significant increase in cell proliferation and a reduction of cell apoptosis. JI-36 increased islet size and glucose-stimulated insulin secretion in isolated rat islets after 48–72 h. At the ultrastructural level, INS-1 cells treated with agonist JI-36 revealed a metabolic active stimulation state with increased cytoplasm. Coincubation with the GHRH antagonist MIA-602 reversed the actions of the agonist JI-36, indicating the specificity of this agonist. In vivo, the function of pancreatic islets was assessed by transplantation of rat islets under the kidney capsule of streptozotocin-induced diabetic non-obese diabetic-severe combined immunodeficiency (NOD-SCID) mice. Islets treated with GHRH agonist JI-36 were able to achieve normoglycemia earlier and more consistently than untreated islets. Furthermore, in contrast to diabetic animals transplanted with untreated islets, insulin response to an i.p. glucose tolerance test (IPGTT) in animals receiving islets treated with agonist Jl-36 was comparable to that of normal healthy mice. In conclusion, our study provides evidence that agonists of GHRH represent a promising pharmacological therapy aimed at promoting islet graft growth and proliferation in diabetic patients.


Endocrinology | 2014

Effects of parathyroid hormone on bone mass, bone strength, and bone regeneration in male rats with type 2 diabetes mellitus.

Christine Hamann; Ann-Kristin Picke; Graeme Campbell; Mariya Balyura; Martina Rauner; Ricardo Bernhardt; Gerd Huber; Michael M. Morlock; Klaus-Peter Günther; Stefan R. Bornstein; Claus-C. Glüer; Barbara Ludwig; Lorenz C. Hofbauer

Type 2 diabetes mellitus (T2DM) is associated with increased skeletal fragility and impaired fracture healing. Intermittent PTH therapy increases bone strength; however, its skeletal and metabolic effects in diabetes are unclear. We assessed whether PTH improves skeletal and metabolic function in rats with T2DM. Subcritical femoral defects were created in diabetic fa/fa and nondiabetic +/+ Zucker Diabetic Fatty (ZDF) rats and internally stabilized. Vehicle or 75 μg/kg/d PTH(1-84) was sc administered over 12 weeks. Skeletal effects were evaluated by μCT, biomechanical testing, histomorphometry, and biochemical markers, and defect regeneration was analyzed by μCT. Glucose homeostasis was assessed using glucose tolerance testing and pancreas histology. In diabetic rats, bone mass was significantly lower in the distal femur and vertebrae, respectively, and increased after PTH treatment by up to 23% in nondiabetic and up to 18% in diabetic rats (P < .0001). Diabetic rats showed 23% lower ultimate strength at the spine (P < .0005), which was increased by PTH by 36% in normal and by 16% in diabetic rats (P < .05). PTH increased the bone formation rate by 3-fold in normal and by 2-fold in diabetic rats and improved defect regeneration in normal and diabetic rats (P < .01). PTH did not affect serum levels of undercarboxylated osteocalcin, glucose tolerance, and islet morphology. PTH partially reversed the adverse skeletal effects of T2DM on bone mass, bone strength, and bone defect repair in rats but did not affect energy metabolism. The positive skeletal effects were generally more pronounced in normal compared with diabetic rats.


Pharmacology & Therapeutics | 2009

An update on preventive and regenerative therapies in diabetes mellitus

Manja Reimann; Ezio Bonifacio; Michele Solimena; Peter Schwarz; Barbara Ludwig; M. Hanefeld; Stefan R. Bornstein

Type 1A (immune-mediated) and type 2 diabetes mellitus are two of the most common severe chronic illnesses, affecting over 230 million people worldwide with an estimated global prevalence of 5.1%. Although type 1 and type 2 diabetes differ greatly in modes of pathogenesis, these illnesses share a common pathology and consequences characterized by loss of functional beta-cell mass and subsequent dysregulation of carbohydrate and lipid metabolism. Since therapy for diabetes and the associated complications poses enormous public health and economic burdens, novel preventive and regenerative therapies have emerged in the past decade with the aim to preserve beta-cell mass and delay the onset of diabetes. The goal of this review is to provide a comprehensive overview of current efforts in the fight against diabetes, and attempts to document all strategies that have emerged in clinical studies within the past 25 years. First, strategies to identify individuals at risk, ranging from whole-genome scans to autoantibody screening, will be discussed. Second, novel approaches to prevent or delay the onset of disease will be covered. Particular focus is given on emerging strategies for individuals at risk for type 1 diabetes that target T-cell regulation and induction of tolerance, while new pharmaceutical concepts in combination with lifestyle interventions are discussed within the scope of type 2 diabetes prevention. Lastly, important efforts to halt disease progression with emphasis on beta-cell regeneration are presented.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Modulation of pancreatic islets-stress axis by hypothalamic releasing hormones and 11β-hydroxysteroid dehydrogenase

Janine Schmid; Barbara Ludwig; Andrew V. Schally; Anja Steffen; Christian G. Ziegler; Norman L. Block; Yassemi Koutmani; Mathias D. Brendel; Katia Karalis; Charmaine J. Simeonovic; Julio Licinio; Monika Ehrhart-Bornstein; Stefan R. Bornstein

Corticotropin-releasing hormone (CRH) and growth hormone-releasing hormone (GHRH), primarily characterized as neuroregulators of the hypothalamic-pituitary-adrenal axis, directly influence tissue-specific receptor-systems for CRH and GHRH in the endocrine pancreas. Here, we demonstrate the expression of mRNA for CRH and CRH-receptor type 1 (CRHR1) and of protein for CRHR1 in rat and human pancreatic islets and rat insulinoma cells. Activation of CRHR1 and GHRH-receptor significantly increased cell proliferation and reduced cell apoptosis. CRH stimulated both cellular content and release of insulin in rat islet and insulinoma cells. At the ultrastructural level, CRHR1 stimulation revealed a more active metabolic state with enlarged mitochondria. Moreover, glucocorticoids that promote glucose production are balanced by both 11b-hydroxysteroid dehydrogenase (11β-HSD) isoforms; 11β-HSD–type-1 and 11β-HSD–type-2. We demonstrated expression of mRNA for 11β-HSD-1 and 11β-HSD-2 and protein for 11β-HSD-1 in rat and human pancreatic islets and insulinoma cells. Quantitative real-time PCR revealed that stimulation of CRHR1 and GHRH-receptor affects the metabolism of insulinoma cells by down-regulating 11β-HSD-1 and up-regulating 11β-HSD-2. The 11β-HSD enzyme activity was analyzed by measuring the production of cortisol from cortisone. Similarly, activation of CRHR1 resulted in reduced cortisol levels, indicating either decreased 11β-HSD-1 enzyme activity or increased 11β-HSD-2 enzyme activity; thus, activation of CRHR1 alters the glucocorticoid balance toward the inactive form. These data indicate that functional receptor systems for hypothalamic-releasing hormone agonists exist within the endocrine pancreas and influence synthesis of insulin and the pancreatic glucocorticoid shuttle. Agonists of CRHR1 and GHRH-receptor, therefore, may play an important role as novel therapeutic tools in the treatment of diabetes mellitus.


Transplantation | 2016

Report from IPITA-TTS Opinion Leaders Meeting on the Future of β-Cell Replacement

Stephen T. Bartlett; James F. Markmann; Paul Johnson; Olle Korsgren; Bernhard J. Hering; David W. Scharp; Thomas W. H. Kay; Jonathan S. Bromberg; Jon S. Odorico; Gordon C. Weir; Nancy D. Bridges; Raja Kandaswamy; Peter Stock; Peter J. Friend; Mitsukazu Gotoh; David K. C. Cooper; Chung Gyu Park; Phillip O'Connell; Cherie L. Stabler; Shinichi Matsumoto; Barbara Ludwig; Pratik Choudhary; Boris P. Kovatchev; Michael R. Rickels; Megan Sykes; Kathryn A Wood; Kristy Kraemer; Albert Hwa; Edward Stanley; Camillo Ricordi

At the time the first pancreas transplant was performed by Kelly and Lillehei in 1966, insulin therapy for diabetes was generally available but administered in a form that is known today as “conventional therapy.”1 In this era, as many as half of all juvenile onset diabetics did not reach the age of 55 years. Early mortality from accelerated cardiovascular disease, renal failure, and hypoglycemia-related events were commonplace. The early low success rate and mortality of pancreas transplantation by comparison were also suboptimal. As will be characterized in the succeeding 8 chapters, the outcome of “best medical therapy” with newer forms of insulin and insulin delivery systems along with dramatically improved outcomes of islet and pancreas transplantation and novel β-cell sources hold great promise for those afflicted.

Collaboration


Dive into the Barbara Ludwig's collaboration.

Top Co-Authors

Avatar

Stefan R. Bornstein

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Anja Steffen

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Stefan Ludwig

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andreas Reichel

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Michele Solimena

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Janine Schmid

Dresden University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge