Network


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

Hotspot


Dive into the research topics where Karl-Henrik Grinnemo is active.

Publication


Featured researches published by Karl-Henrik Grinnemo.


The Lancet | 2011

Tracheobronchial transplantation with a stem-cell-seeded bioartificial nanocomposite: a proof-of-concept study

Philipp Jungebluth; Evren Alici; Silvia Baiguera; Katarina Le Blanc; Pontus Blomberg; Béla Bozóky; Claire Crowley; Oskar Einarsson; Karl-Henrik Grinnemo; Tomas Gudbjartsson; Sylvie Le Guyader; Gert Henriksson; Ola Hermanson; Jan Erik Juto; Bertil Leidner; Tobias Lilja; Jan Liska; Tom Luedde; Vanessa Lundin; Guido Moll; Bo Nilsson; Christoph Roderburg; Staffan Strömblad; Tolga Sutlu; Ana I. Teixeira; Emma Watz; Alexander M. Seifalian; Paolo Macchiarini

BACKGROUND Tracheal tumours can be surgically resected but most are an inoperable size at the time of diagnosis; therefore, new therapeutic options are needed. We report the clinical transplantation of the tracheobronchial airway with a stem-cell-seeded bioartificial nanocomposite. METHODS A 36-year-old male patient, previously treated with debulking surgery and radiation therapy, presented with recurrent primary cancer of the distal trachea and main bronchi. After complete tumour resection, the airway was replaced with a tailored bioartificial nanocomposite previously seeded with autologous bone-marrow mononuclear cells via a bioreactor for 36 h. Postoperative granulocyte colony-stimulating factor filgrastim (10 μg/kg) and epoetin beta (40,000 UI) were given over 14 days. We undertook flow cytometry, scanning electron microscopy, confocal microscopy epigenetics, multiplex, miRNA, and gene expression analyses. FINDINGS We noted an extracellular matrix-like coating and proliferating cells including a CD105+ subpopulation in the scaffold after the reseeding and bioreactor process. There were no major complications, and the patient was asymptomatic and tumour free 5 months after transplantation. The bioartificial nanocomposite has patent anastomoses, lined with a vascularised neomucosa, and was partly covered by nearly healthy epithelium. Postoperatively, we detected a mobilisation of peripheral cells displaying increased mesenchymal stromal cell phenotype, and upregulation of epoetin receptors, antiapoptotic genes, and miR-34 and miR-449 biomarkers. These findings, together with increased levels of regenerative-associated plasma factors, strongly suggest stem-cell homing and cell-mediated wound repair, extracellular matrix remodelling, and neovascularisation of the graft. INTERPRETATION Tailor-made bioartificial scaffolds can be used to replace complex airway defects. The bioreactor reseeding process and pharmacological-induced site-specific and graft-specific regeneration and tissue protection are key factors for successful clinical outcome. FUNDING European Commission, Knut and Alice Wallenberg Foundation, Swedish Research Council, StratRegen, Vinnova Foundation, Radiumhemmet, Clinigene EU Network of Excellence, Swedish Cancer Society, Centre for Biosciences (The Live Cell imaging Unit), and UCL Business.


Nature Communications | 2014

Clonal culturing of human embryonic stem cells on laminin-521/E-cadherin matrix in defined and xeno-free environment

Sergey Rodin; Liselotte Antonsson; Colin Niaudet; Oscar E. Simonson; Elina Salmela; Emil M. Hansson; Anna Domogatskaya; Zhijie Xiao; Pauliina Damdimopoulou; Mona Sheikhi; José Inzunza; Ann-Sofie Nilsson; Duncan Baker; Raoul Kuiper; Yi Sun; Elisabeth Blennow; Magnus Nordenskjöld; Karl-Henrik Grinnemo; Juha Kere; Christer Betsholtz; Outi Hovatta; Karl Tryggvason

Lack of robust methods for establishment and expansion of pluripotent human embryonic stem (hES) cells still hampers development of cell therapy. Laminins (LN) are a family of highly cell-type specific basement membrane proteins important for cell adhesion, differentiation, migration and phenotype stability. Here we produce and isolate a human recombinant LN-521 isoform and develop a cell culture matrix containing LN-521 and E-cadherin, which both localize to stem cell niches in vivo. This matrix allows clonal derivation, clonal survival and long-term self-renewal of hES cells under completely chemically defined and xeno-free conditions without ROCK inhibitors. Neither LN-521 nor E-cadherin alone enable clonal survival of hES cells. The LN-521/E-cadherin matrix allows hES cell line derivation from blastocyst inner cell mass and single blastomere cells without a need to destroy the embryo. This method can facilitate the generation of hES cell lines for development of different cell types for regenerative medicine purposes.


Reproductive Biomedicine Online | 2006

Human embryonic stem cells are immunogenic in allogeneic and xenogeneic settings

Karl-Henrik Grinnemo; Makiko Kumagai-Braesch; Agneta Mânsson-Broberg; Heli Skottman; Xiaojin Hao; Anwar J. Siddiqui; Agneta Andersson; Anne-Marie Strömberg; Riita Lahesmaa; Outi Hovatta; Christer Sylvén; Matthias Corbascio; Göran Dellgren

Recent studies have suggested that human embryonic stem cells (HESC) are immune-privileged and may thereby circumvent rejection. The expression of immunologically active molecules was studied by DNA microarray analysis and by flow cytometry. HESC were transplanted into immunologically competent mice and traced by fluorescence in-situ hybridization (FISH) and immunohistochemistry. The ability of HESC to directly and indirectly induce immune responses in CD4+ T-cells from naive and transplanted mice was studied. Their ability to induce immune responses of human CD4+ T-cells, when cultured in the presence of dendritic cells (DC) syngeneic to responder T-cells, was also analysed. HESC demonstrated expression of HLA class I and HLA class II genes, but the cell surface expression of HLA class II molecules was low even after incubation with IFNgamma. In wild-type mice, HESC could be demonstrated by FISH until 3 days after transplantation and were surrounded by heavy infiltrates of T-cells and macrophages. HESC induced a similar immune response as human fibroblast cells (HFib) on naive and immunized T-cells, both directly and in the presence of syngeneic DC. A similar response was observed in the allogeneic setting. It is concluded that HESC are immunologically inert and do not inhibit immune responses during direct or indirect antigen presentation, and they were acutely rejected in a xenogeneic setting.


Cell and Tissue Research | 2008

Immunogenicity of human embryonic stem cells

Karl-Henrik Grinnemo; Christer Sylvén; Outi Hovatta; Göran Dellgren; Matthias Corbascio

Human embryonic stem cells (HESC) are pluripotent stem cells isolated from the inner cell mass of human blastocysts. With the first successful culturing of HESC, a new era of regenerative medicine was born. HESC can differentiate into almost any cell type and, in the future, might replace solid organ transplantation and even be used to treat progressive degenerative diseases such as Parkinson’s disease. Although this sounds promising, certain obstacles remain with regard to their clinical use, such as culturing HESC under well-defined conditions without exposure to animal proteins, the risk of teratoma development and finally the avoidance of immune rejection. In this review, we discuss the immunological properties of HESC and various strategic solutions to circumvent immune rejection, such as stem cell banking, somatic cell nuclear transfer and the induction of tolerance by co-stimulation blockade and mixed chimerism.


Annals of Medicine | 2006

Human mesenchymal stem cells do not differentiate into cardiomyocytes in a cardiac ischemic xenomodel

Karl-Henrik Grinnemo; Agneta Månsson-Broberg; Katarina LeBlanc; Matthias Corbascio; Eva Wärdell; Anwar J. Siddiqui; Xiaojin Hao; Christer Sylvén; Göran Dellgren

AIM. As the capability of human mesenchymal stem cells (hMSC) to engraft, differentiate and improve myocardial function cannot be studied in humans, exploration was performed in a xenomodel. METHODS. The rats were divided into three groups depending on the type of rats used (Rowett nude (RNU) or Fischer rats +/− immunosuppression). Different groups were treated with intramyocardial injection of hMSC (1–2 million) either directly or three days after ligation of the left anterior descending artery (LAD). Myocardial function was investigated by echocardiography. The hMSC were identified with fluorescence in situ hybridization and myocardial differentiation was assessed by immunohistochemistry. RESULTS. When hMSC were injected directly after LAD ligation they could be identified in half (8/16) of the RNU rats (without immunosuppression) at 4 weeks. When injected 3 days after LAD ligation in immunosuppressed RNU rats they were identified in all (6/6) rats at 6 weeks. The surviving hMSC showed signs of differentiation into fibroblasts. No cardiomyocyte differentiation was observed. There was no difference in myocardial function in treated animals compared to controls. CONCLUSIONS. The hMSC survived in this xenomodel up to 6 weeks. However, hMSC required implantation into immunoincompetent animals as well as immunosuppression to survive, indicating that these cells are otherwise rejected. Furthermore, these cells did not differentiate into cardiomyocytes nor did they improve heart function in this xenomodel.


Circulation Research | 2013

Local Control of Nuclear Calcium Signaling in Cardiac Myocytes by Perinuclear Microdomains of Sarcolemmal Insulin-Like Growth Factor 1 Receptors

Cristián Ibarra; Jose Miguel Vicencio; Manuel Estrada; Yingbo Lin; Paola Rocco; Paola Rebellato; Juan Pablo Muñoz; Jaime García-Prieto; Andrew F.G. Quest; Mario Chiong; Sean M. Davidson; Ivana Bulatovic; Karl-Henrik Grinnemo; Olle Larsson; Per Uhlén; Enrique Jaimovich; Sergio Lavandero

Rationale: The ability of a cell to independently regulate nuclear and cytosolic Ca2+ signaling is currently attributed to the differential distribution of inositol 1,4,5-trisphosphate receptor channel isoforms in the nucleoplasmic versus the endoplasmic reticulum. In cardiac myocytes, T-tubules confer the necessary compartmentation of Ca2+ signals, which allows sarcomere contraction in response to plasma membrane depolarization, but whether there is a similar structure tunneling extracellular stimulation to control nuclear Ca2+ signals locally has not been explored. Objective: To study the role of perinuclear sarcolemma in selective nuclear Ca2+ signaling. Methods and Results: We report here that insulin-like growth factor 1 triggers a fast and independent nuclear Ca2+ signal in neonatal rat cardiac myocytes, human embryonic cardiac myocytes, and adult rat cardiac myocytes. This fast and localized response is achieved by activation of insulin-like growth factor 1 receptor signaling complexes present in perinuclear invaginations of the plasma membrane. The perinuclear insulin-like growth factor 1 receptor pool connects extracellular stimulation to local activation of nuclear Ca2+ signaling and transcriptional upregulation through the perinuclear hydrolysis of phosphatidylinositol 4,5-biphosphate inositol 1,4,5-trisphosphate production, nuclear Ca2+ release, and activation of the transcription factor myocyte-enhancing factor 2C. Genetically engineered Ca2+ buffers—parvalbumin—with cytosolic or nuclear localization demonstrated that the nuclear Ca2+ handling system is physically and functionally segregated from the cytosolic Ca2+ signaling machinery. Conclusions: These data reveal the existence of an inositol 1,4,5-trisphosphate–dependent nuclear Ca2+ toolkit located in direct apposition to the cell surface, which allows the local control of rapid and independent activation of nuclear Ca2+ signaling in response to an extracellular ligand.


Stem Cells and Development | 2010

Islet-1 cells are cardiac progenitors present during the entire lifespan: from the embryonic stage to adulthood.

Rami Genead; Christian Danielsson; Agneta Andersson; Matthias Corbascio; Anders Franco-Cereceda; Christer Sylvén; Karl-Henrik Grinnemo

The aim of this study was to longitudinally characterize the distribution of cells actively expressing the progenitor transcription factor islet-1 (Isl1+) during the embryonic life, the postnatal period, and adulthood. In this study, we have used direct immunohistochemical staining toward the protein Isl1 in a longitudinal rat model. Cells actively expressing Isl1 were traced in embryos from gestational day (GD) 11 until adulthood. In early cardiac development (GD 11), the Isl1+ progenitors were located in a greater abundance in the paracardiac regions, areas suggested to be the second heart field. To a lesser extent, Isl1+ cells were present within the bulbotruncal region and the truncus arteriosus. During the following days until GD 15, the Isl1+ cells were mainly observed at the proximal outflow tract (OFT) and at the inflow area of the right atrium. No Isl1+ cells were detected in the left ventricle. Compared with GD 11, more Isl1+ cells seemed to co-express cardiomyocyte markers and a minority of the Isl1+ cells was undifferentiated. Unexpectedly, only few undifferentiated Isl1+ cells were Ki67+ while a lot of TnT+ cardiomyocytes were proliferating in the ventricles. After birth, immature Isl1+ cells were still present in the OFT where they resided until adulthood. Our data suggest that during embryogenesis, Isl1+ cells migrate from extracardiac regions into the proximal part of the heart, proliferating and giving rise to cardioblasts. Unexpectedly, only a minority of the Isl1+ cells while a majority of ventricular cardiomyocytes were proliferating. The Isl1+ cell pool persists into adulthood, which might open up new strategies to repair damaged myocardium.


Circulation Research | 2012

Local Control of Nuclear Ca2+ Signalling in Cardiac Myocytes by Perinuclear Microdomains of Sarcolemmal IGF-1 Receptors

Cristián Ibarra; Jose Miguel Vicencio; Manuel Estrada; Yingbo Lin; Paola Rocco; Paola Rebellato; Juan Pablo Muñoz; Jaime García-Prieto; Andrew F.G. Quest; Mario Chiong; Sean M. Davidson; Ivana Bulatovic; Karl-Henrik Grinnemo; Olle Larsson; Per Uhlén; Enrique Jaimovich; Sergio Lavandero

Rationale: The ability of a cell to independently regulate nuclear and cytosolic Ca2+ signaling is currently attributed to the differential distribution of inositol 1,4,5-trisphosphate receptor channel isoforms in the nucleoplasmic versus the endoplasmic reticulum. In cardiac myocytes, T-tubules confer the necessary compartmentation of Ca2+ signals, which allows sarcomere contraction in response to plasma membrane depolarization, but whether there is a similar structure tunneling extracellular stimulation to control nuclear Ca2+ signals locally has not been explored. Objective: To study the role of perinuclear sarcolemma in selective nuclear Ca2+ signaling. Methods and Results: We report here that insulin-like growth factor 1 triggers a fast and independent nuclear Ca2+ signal in neonatal rat cardiac myocytes, human embryonic cardiac myocytes, and adult rat cardiac myocytes. This fast and localized response is achieved by activation of insulin-like growth factor 1 receptor signaling complexes present in perinuclear invaginations of the plasma membrane. The perinuclear insulin-like growth factor 1 receptor pool connects extracellular stimulation to local activation of nuclear Ca2+ signaling and transcriptional upregulation through the perinuclear hydrolysis of phosphatidylinositol 4,5-biphosphate inositol 1,4,5-trisphosphate production, nuclear Ca2+ release, and activation of the transcription factor myocyte-enhancing factor 2C. Genetically engineered Ca2+ buffers—parvalbumin—with cytosolic or nuclear localization demonstrated that the nuclear Ca2+ handling system is physically and functionally segregated from the cytosolic Ca2+ signaling machinery. Conclusions: These data reveal the existence of an inositol 1,4,5-trisphosphate–dependent nuclear Ca2+ toolkit located in direct apposition to the cell surface, which allows the local control of rapid and independent activation of nuclear Ca2+ signaling in response to an extracellular ligand.


Stem Cells | 2008

Costimulation Blockade Induces Tolerance to HESC Transplanted to the Testis and Induces Regulatory T‐Cells to HESC Transplanted into the Heart

Karl-Henrik Grinnemo; Rami Genead; Makiko Kumagai-Braesch; Agneta Andersson; Christian Danielsson; Agneta Månsson-Broberg; Göran Dellgren; Anne-Marie Strömberg; Henrik Ekberg; Outi Hovatta; Christer Sylvén; Matthias Corbascio

In order to study the ability of costimulation blockade to induce tolerance to human embryonic stem cells (HESC), severe combined immunodeficient (SCID), and immunocompetent C57BL/6 mice treated with costimulation blockade received intratesticular and intramyocardial HESC transplants. All SCID mice with intratesticular HESC transplants developed teratoma. When SCID mice were transplanted intramyocardially, only two of five mice developed teratoma‐like tumors. C57BL/6 mice transplanted intratesticularly and treated with costimulation blockade all developed teratoma and were surrounded by CD4+CD25+Foxp3+ T‐cells, while isotype control treated recipients rejected their grafts. Most C57BL/6 mice transplanted intramyocardially and treated with costimulation blockade demonstrated lymphocytic infiltrates 1 month after transplantation, whereas one maintained its graft. Isolation of regulatory T‐cells from intramyocardial transplanted recipients treated with costimulation blockade demonstrated specificity toward undifferentiated HESC and down‐regulated naive T‐cell activation toward HESC. These results demonstrate that costimulation blockade is sufficiently robust to induce tolerance to HESC in the immune‐privileged environment of the testis. HESC specific regulatory T‐cells developed to HESC transplanted to the heart and the success of transplantation was similar to that seen in SCID mice.


Biochemical and Biophysical Research Communications | 2008

Modulation of ephrinB2 leads to increased angiogenesis in ischemic myocardium and endothelial cell proliferation

Agneta Månsson-Broberg; Anwar J. Siddiqui; Maria Genander; Karl-Henrik Grinnemo; Xiaojin Hao; Agneta Andersson; Eva Wärdell; Christer Sylvén; Matthias Corbascio

Eph/ephrin signaling is pivotal in prenatal angiogenesis while its potential role in postnatal angiogenesis largely remains to be explored. Therefore its putative angiogenic and therapeutic effects were explored in endothelium and in myocardial ischemia. In culture of human aortic endothelial cells the fusion protein ephrinB2-Fc induced cell proliferation (p<0.0005) and in the murine aortic ring model ephrinB2-Fc induced increased sprouting (p<0.05). Myocardial infarction was induced by ligation of the left anterior descending artery in mouse. During the following 2 weeks mRNA of the receptor/ligand pair EphB4/ephrinB2 was expressed dichotomously (p<0.05) and other Eph/ephrin pairs were expressed to a lesser degree. Twenty-four hours after intraperitoneal administration of ephrinB2-Fc it was detected in abundance throughout the myocardium along capillaries, showing signs of increased mitosis. After 4 weeks the capillary density was increased 28% in the periinfarcted area (p<0.05) to a level not different from healthy regions of the heart where no change was observed. These results implicate that EphB4/ephrinB2 is an important signaling pathway in ischemic heart disease and its modulation may induce therapeutic angiogenesis.

Collaboration


Dive into the Karl-Henrik Grinnemo's collaboration.

Top Co-Authors

Avatar

Christer Sylvén

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Agneta Månsson-Broberg

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Matthias Corbascio

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Eva Wärdell

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anwar J. Siddiqui

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Xiaojin Hao

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Agneta Andersson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Peter Svenarud

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Rami Genead

Karolinska University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge