Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Heliyon ; 7(2): e06129, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33644446

ABSTRACT

BACKGROUND: Biological organ engineering is a novel experimental approach to generate functional liver grafts by decellularization and repopulation. Currently, healthy organs of small or large animals and human organs with preexisting liver diseases are used to optimize decellularization and repopulation.However, the effects of morphological changes on allo- and xenogeneic cell-scaffold interactions during repopulation procedure, e.g., using scaffold-sections, are unknown. We present a sequential morphological workflow to identify murine liver scaffold-sections with well-preserved microarchitecture. METHODS: Native livers (CONT, n = 9) and livers with experimentally induced pathologies (hepatics steatosis: STEA, n = 7; hepatic fibrosis induced by bile duct ligation: BDL, n = 9; nodular regenerative hyperplasia induced by 90% partial hepatectomy: PH, n = 8) were decellularized using SDS and Triton X-100 to generate cell-free scaffolds. Scaffold-sections were assessed using a sequential morphological workflow consisting of macroscopic, microscopic and morphological evaluation: (1) The scaffold was evaluated by a macroscopic decellularization score. (2) Regions without visible tissue remnants were localized for sampling and histological processing. Subsequent microscopical examination served to identify tissue samples without cell remnants. (3) Only cell-free tissue sections were subjected to detailed liver-specific morphological assessment using a histological and immunohistochemical decellularization score. RESULTS: Decellularization was feasible in 33 livers, which were subjected to the sequential morphological workflow. In 11 of 33 scaffolds we achieved a good macroscopic decellularization result (CONT: 3 scaffolds; STEA: 3 scaffolds; BDL: 3 scaffolds; PH: 2 scaffolds). The microscopic assessment resulted in the selection of 88 cell-free tissue sections (CONT: 15 sections; STEA: 38 sections; BDL: 30 sections; PH: 5 sections). In 27 of those sections we obtained a good histological decellularization result (CONT: 3 sections; STEA: 6 sections; BDL: 17 sections; PH: 1 section). All experimental groups contained sections with a good immunohistochemical decellularization result (CONT: 6 sections; STEA: 5 sections; BDL: 4 sections; PH: 1 section). DISCUSSION: Decellularization was possible in all experimental groups, irrespectively of the underlying morphological alteration. Furthermore, our proposed sequential morphological workflow was suitable to detect tissue sections with well-preserved hepatic microarchitecture, as needed for further repopulation experiments.

2.
Transplant Proc ; 52(1): 406-413, 2020.
Article in English | MEDLINE | ID: mdl-31911055

ABSTRACT

BACKGROUND: Subcutaneous ovarian transplantation has recently begun receiving increased attention. Fourteen days after transplantation is used as an important time point for assessing the recovery of ovarian function. The goal of this study is to determine the expression of apoptotic genes in the ovary at this time. METHODS: This study investigated follicle development and the expression of 3 apoptosis genes (Bax, Bcl2, and P53) after mouse ovaries were transplanted. Seven-week-old mouse ovaries were autologously transplanted into back muscle. The ovaries were harvested on day 14, morphology was observed by hematoxylin and eosin staining, and the distribution of 3 proteins was observed by immunohistochemistry. TUNEL staining showed where apoptosis occurred in the ovary. Finally, RT-PCR/Western blotting was used to analyze the differential expression of mRNA/proteins between the transplantation group and the control group. RESULTS: The results revealed follicles at different stages at the edge of the grafts. In immunohistochemical experiments, BAX, BCL2, and P53 were found to be extensively expressed in the transplant group and the control group. P53 was strongly expressed in the medulla of transplanted ovaries. Bax was strongly expressed in the antral follicles of both groups. The results were consistent with the results of the TUNEL experiments. Three genes (Bax, Bcl2, and P53) were downregulated in the transplanted groups. The results showed that significant differences were detected in Bax and P53 mRNA expression levels between the transplanted groups and the control group (P < .01). Bcl2 expression was not significantly different, but the Bax/Bcl2 ratio increased. The results of the protein experiments were the same. CONCLUSION: P53 may downregulate Bax in the early stage of transplantation. Follicle growth and atresia were regulated through modulation of Bcl2- and Bax-mediated apoptotic pathways in heterotopic ovarian transplantation.


Subject(s)
Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Ovary/metabolism , Ovary/pathology , Ovary/transplantation , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/physiology , Female , Mice , Mice, Inbred C57BL , Transplantation, Autologous , bcl-2-Associated X Protein/metabolism
3.
Int J Mol Sci ; 18(1)2017 Jan 16.
Article in English | MEDLINE | ID: mdl-28275217

ABSTRACT

Ischemia/reperfusion injury (IRI) occurs inevitably in liver transplantations and frequently during major resections, and can lead to liver dysfunction as well as systemic disorders. High-mobility group box 1 (HMGB1) plays a pathogenic role in hepatic IRI. In the normal liver, HMGB1 is located in the nucleus of hepatocytes; after ischemia reperfusion, it translocates to the cytoplasm and it is further released to the extracellular space. Unlike the well-explored functions of nuclear and extracellular HMGB1, the role of cytoplasmic HMGB1 in hepatic IRI remains elusive. We hypothesized that cytoplasmic HMGB1 interacts with binding proteins involved in the hepatocellular response to IRI. In this study, binding proteins of cytoplasmic HMGB1 during hepatic IRI were identified. Liver tissues from rats with warm ischemia reperfusion (WI/R) injury and from normal rats were subjected to cytoplasmic protein extraction. Co-immunoprecipitation using these protein extracts was performed to enrich HMGB1-protein complexes. To separate and identify the immunoprecipitated proteins in eluates, 2-dimensional electrophoresis and subsequent mass spectrometry detection were performed. Two of the identified proteins were verified using Western blotting: betaine-homocysteine S-methyltransferase 1 (BHMT) and cystathionine γ-lyase (CTH). Therefore, our results revealed the binding of HMGB1 to BHMT and CTH in cytoplasm during hepatic WI/R. This finding may help to better understand the cellular response to IRI in the liver and to identify novel molecular targets for reducing ischemic injury.


Subject(s)
Cytoplasm/metabolism , HMGB1 Protein/metabolism , Hepatocytes/metabolism , Reperfusion Injury/metabolism , Animals , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Liver/metabolism , Male , Mass Spectrometry , Protein Binding , Rats, Inbred Lew , Warm Ischemia
4.
J Vis Exp ; (115)2016 09 13.
Article in English | MEDLINE | ID: mdl-27685096

ABSTRACT

A modified silicone injection procedure was used for visualization of the hepatic vascular tree. This procedure consisted of in-vivo injection of the silicone compound, via a 26 G catheter, into the portal or hepatic vein. After silicone injection, organs were explanted and prepared for ex-vivo micro-CT (µCT) scanning. The silicone injection procedure is technically challenging. Achieving a successful outcome requires extensive microsurgical experience from the surgeon. One of the challenges of this procedure involves determining the adequate perfusion rate for the silicone compound. The perfusion rate for the silicone compound needs to be defined based on the hemodynamic of the vascular system of interest. Inappropriate perfusion rate can lead to an incomplete perfusion, artificial dilation and rupturing of vascular trees. The 3D reconstruction of the vascular system was based on CT scans and was achieved using preclinical software such as HepaVision. The quality of the reconstructed vascular tree was directly related to the quality of silicone perfusion. Subsequently computed vascular parameters indicative of vascular growth, such as total vascular volume, were calculated based on the vascular reconstructions. Contrasting the vascular tree with silicone allowed for subsequent histological work-up of the specimen after µCT scanning. The specimen can be subjected to serial sectioning, histological analysis and whole slide scanning, and thereafter to 3D reconstruction of the vascular trees based on histological images. This is the prerequisite for the detection of molecular events and their distribution with respect to the vascular tree. This modified silicone injection procedure can also be used to visualize and reconstruct the vascular systems of other organs. This technique has the potential to be extensively applied to studies concerning vascular anatomy and growth in various animal and disease models.


Subject(s)
Hepatic Veins/diagnostic imaging , Liver Regeneration/physiology , Liver/blood supply , Portal Vein/diagnostic imaging , Regeneration/physiology , Animals , Contrast Media/administration & dosage , Female , Hepatectomy , Hepatic Veins/physiology , Liver/surgery , Male , Mice , Portal Vein/physiology , Silicones/administration & dosage , Software , Tomography, X-Ray Computed/methods
5.
PLoS One ; 11(8): e0160581, 2016.
Article in English | MEDLINE | ID: mdl-27494255

ABSTRACT

BACKGROUND: Liver regeneration consists of cellular proliferation leading to parenchymal and vascular growth. This study complements previous studies on cellular proliferation and weight recovery by (1) quantitatively describing parenchymal and vascular regeneration, and (2) determining their relationship. Both together are needed to (3) characterize the underlying growth pattern. METHODS: Specimens were created by injecting a polymerizing contrast agent in either portal or hepatic vein in normal or regenerating livers after 70% partial hepatectomy. 3D image data were obtained through micro-CT scanning. Parenchymal growth was assessed by determining weight and volume of the regenerating liver. Vascular growth was described by manually determined circumscribed parameters (maximal vessel length and radius of right inferior portal/hepatic vein), automatically determined cumulative parameters (total edge length and total vascular volume), and parameters describing vascular density (total edge length/volume, vascular volume fraction). The growth pattern was explored by comparing the relative increase of these parameters to the increase expected in case of isotropic expansion. RESULTS: Liver volume recovery paralleled weight recovery and reached 90% of the original liver volume within 7 days. Comparing radius-related vascular parameters immediately after surgical resection and after virtual resection in-silico revealed a slight increase, possibly reflecting the effect of resection-induced portal hyperperfusion. Comparing length-related parameters between post-operative day 7 and after virtual resection showed similar vascular growth in both vascular systems investigated. In contrast, radius-related parameters increased slightly more in the portal vein. Despite the seemingly homogeneous 3D growth, the observed vascular parameters were not compatible with the hypothesis of isotropic expansion of liver parenchyma and vascular structures. CONCLUSION: We present an approach for the quantitative analysis of the vascular systems of regenerating mouse livers. We applied this technique for assessing the hepatic growth pattern. Prospectively, this approach can be used to investigate hepatic vascular regeneration under different conditions.


Subject(s)
Hepatic Artery/cytology , Hepatic Veins/cytology , Liver Regeneration/physiology , Liver/cytology , Parenchymal Tissue/cytology , Animals , Hepatectomy , Hepatic Artery/diagnostic imaging , Hepatic Veins/diagnostic imaging , Imaging, Three-Dimensional , Liver/blood supply , Liver/diagnostic imaging , Liver/surgery , Male , Mice , Mice, Inbred C57BL , Parenchymal Tissue/diagnostic imaging , Tomography, X-Ray Computed
6.
Eur Surg Res ; 57(3-4): 224-239, 2016.
Article in English | MEDLINE | ID: mdl-27459202

ABSTRACT

BACKGROUND: Organ engineering is a new innovative strategy to cope with two problems: the need for physiological models for pharmacological research and donor organs for transplantation. A functional scaffold is generated from explanted organs by removing all cells (decellularization) by perfusing the organ with ionic or nonionic detergents via the vascular system. Subsequently the acellular scaffold is reseeded with organ-specific cells (repopulation) to generate a functional organ. SUMMARY: This review gives an overview of the state of the art describing the decellularization process, the subsequent quality assessment, the repopulation techniques and the functional assessment. It emphasizes the use of scaffolds as matrix for culturing human liver cells for drug testing. Further, it highlights the techniques for transplanting these engineered scaffolds in allogeneic or xenogeneic animals in order to test their biocompatibility and use as organ grafts. Key Messages: The first issue is the so-called decellularization, which is best explored and resulted in a multitude of different protocols. The most promising approach seems to be the combination of pulsatile perfusion of the liver with Triton X-100 and SDS via hepatic artery and portal vein. Widely accepted parameters of quality control include the quantitative assessment of the DNA content and the visualization of eventually remaining nuclei confirmed by HE staining. Investigations regarding the composition of the extracellular matrix focused on histological determination of laminin, collagen, fibronectin and elastin and remained qualitatively. Repopulation is the second issue which is addressed. Selection of the most suitable cell type is a highly controversial topic. Currently, the highest potential is seen for progenitor and stem cells. Cells are infused into the scaffold and cultured under static conditions or in a bioreactor allowing dynamic perfusion of the scaffold. The quality of repopulation is mainly assessed by routine histology and basic functional assays. These promising results prompted to consider the use of a liver scaffold repopulated with human cells for pharmacological research. Transplantation of the (repopulated) scaffold is the third topic which is not yet widely addressed. Few studies report the heterotopic transplantation of repopulated liver tissue without vascular anastomosis. Even fewer studies deal with the heterotopic transplantation of a scaffold or a repopulated liver lobe. However, observation time was still limited to hours, and long-term graft survival has not been reported yet. These exciting results emphasize the potential of this new and promising strategy to create physiological models for pharmacological research and to generate liver grafts for the transplant community to treat organ failure. However, the scientific need for further development in the field of liver engineering is still tremendous.


Subject(s)
Liver Transplantation , Liver/cytology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Bioreactors , Cells, Cultured , Humans , Liver/physiology
7.
Surgery ; 159(5): 1299-307, 2016 May.
Article in English | MEDLINE | ID: mdl-26879073

ABSTRACT

BACKGROUND: We adapted the anatomically oriented parenchyma-preserving resection technique for associating liver partition with portal vein ligation (PVL) for staged hepatectomy (ALPPS) in rats and examined the role of revascularization in intrahepatic size regulation. METHODS: We performed the procedures based on anatomic study. The ALPPS procedure consisted of a 70% PVL (occluding the left median, left lateral, and right lobes), parenchymal transection (median lobe) and partial (10%) hepatectomy (PHx; caudate lobe). The transection effect was evaluated by measuring the extent of hepatic atrophy or regeneration of individual liver lobes in the ALPPS and control groups (70% PVL and 10% PHx without transection). The survival rates after stage II resection and collateral formation within the portal vein system was examined. RESULTS: Anatomic study revealed a close spatial relationship between the demarcation line and the middle median hepatic vein. This enabled placing the transection plane without injuring the hepatic vein. Transection was achieved via stepwise clamping, followed by 2-3 parenchyma-preserving piercing sutures on both sides of the clamp. Ligated liver lobes atrophy was significantly enhanced after ALPPS compared with the control group. In contrast, both a significantly greater relative weight of the regenerated lobe and proliferation index on the first postoperative day were observed. All animals tolerated stage II-resection without complications. Portoportal collaterals were only observed in the control group. CONCLUSION: We developed an anatomically precise technique for parenchymal transection. The lack of a dense vascular network between the portalized and deportalized lobes may play an important role in accelerating regeneration and atrophy augmentation.


Subject(s)
Hepatectomy/methods , Liver Regeneration , Liver/surgery , Models, Animal , Portal Vein/surgery , Animals , Ligation , Liver/blood supply , Liver/physiology , Male , Rats , Rats, Inbred Lew
8.
PLoS One ; 10(11): e0141798, 2015.
Article in English | MEDLINE | ID: mdl-26618494

ABSTRACT

INTRODUCTION: The intra-hepatic vascular anatomy in rodents, its variations and corresponding supplying and draining territories in respect to the lobar structure of the liver have not been described. We performed a detailed anatomical imaging study in rats and mice to allow for further refinement of experimental surgical approaches. METHODS: LEWIS-Rats and C57Bl/6N-Mice were subjected to ex-vivo imaging using µCT. The image data were used for semi-automated segmentation to extract the hepatic vascular tree as prerequisite for 3D visualization. The underlying vascular anatomy was reconstructed, analysed and used for determining hepatic vascular territories. RESULTS: The four major liver lobes have their own lobar portal supply and hepatic drainage territories. In contrast, the paracaval liver is supplied by various small branches from right and caudate portal veins and drains directly into the vena cava. Variations in hepatic vascular anatomy were observed in terms of branching pattern and distance of branches to each other. The portal vein anatomy is more variable than the hepatic vein anatomy. Surgically relevant variations were primarily observed in portal venous supply. CONCLUSIONS: For the first time the key variations of intrahepatic vascular anatomy in mice and rats and their surgical implications were described. We showed that lobar borders of the liver do not always match vascular territorial borders. These findings are of importance for the design of new surgical procedures and for understanding eventual complications following hepatic surgery.


Subject(s)
Liver/blood supply , Mice, Inbred C57BL/anatomy & histology , Rats, Inbred Lew/anatomy & histology , Animals , Hepatic Artery/anatomy & histology , Hepatic Artery/surgery , Hepatic Veins/anatomy & histology , Hepatic Veins/surgery , Liver/anatomy & histology , Liver/surgery , Mice , Mice, Inbred C57BL/surgery , Microvessels/anatomy & histology , Microvessels/surgery , Rats , Rats, Inbred Lew/surgery
9.
PLoS One ; 10(8): e0135049, 2015.
Article in English | MEDLINE | ID: mdl-26271079

ABSTRACT

Ischemia is the first mechanism that provokes the loss of follicles in ovarian grafts over the long term. In whole ovarian transplantation, it remains unknown, however, how changes in follicular development are influenced by short-term ischemia. Fresh whole ovarian orthotopic auto-transplantation was performed in rabbits with 45 min ischemia, and the impact of ischemia on follicular survival and development status was evaluated at different time-points (1 day, 3 days, 1 week, 2 weeks and 1 month). Assessment of follicular quantity and morphology was carried out via histologic analysis. Follicle proliferating status was evidenced by immunostaining with proliferating cell nuclear antigen (PCNA), and the Hedgehog signaling pathway (Patched and Gli); was verified via TUNEL assay. Quantitative PCR was carried out to quantify the mRNA of target genes including PCNA, Patched, Gli, Caspase 3, Bax, and Bcl-2. Compared with its contralateral fresh controls, the morphology, proliferation and apoptosis of the follicles in the grafts showed no significant differences and most primordial follicles were quiescent. However, morphology and proliferation status were significantly decreased 1 week after grafting, in comparison with the longitudinal grafting time. Patched and Gli in the Hedgehog signaling pathway were activated in only the follicles of the grafts. Short-term ischemia slightly impacts follicular survival and development status in whole ovarian grafting. Receiving intervention in the first week post-transplantation might be helpful.


Subject(s)
Graft Survival , Ischemia/pathology , Ovarian Follicle/blood supply , Ovarian Follicle/physiology , Ovary/blood supply , Ovary/transplantation , Animals , Cell Count , Cell Shape , Cell Survival , Female , Ischemia/metabolism , Ovarian Follicle/pathology , Ovary/metabolism , Ovary/pathology , Proliferating Cell Nuclear Antigen/metabolism , Rabbits , Transplantation, Heterologous
10.
J Vis Exp ; (92): e51955, 2014 Oct 04.
Article in English | MEDLINE | ID: mdl-25350047

ABSTRACT

The use of mouse models in experimental research is of enormous importance for the study of hepatic physiology and pathophysiological disturbances. However, due to the small size of the mouse, technical details of the intraoperative monitoring procedure suitable for the mouse were rarely described. Previously we have reported a monitoring procedure to obtain hemodynamic parameters for rats. Now, we adapted the procedure to acquire systemic and hepatic hemodynamic parameters in mice, a species ten-fold smaller than rats. This film demonstrates the instrumentation of the animals as well as the data acquisition process needed to assess systemic and hepatic hemodynamics in mice. Vital parameters, including body temperature, respiratory rate and heart rate were recorded throughout the whole procedure. Systemic hemodynamic parameters consist of carotid artery pressure (CAP) and central venous pressure (CVP). Hepatic perfusion parameters include portal vein pressure (PVP), portal flow rate as well as the flow rate of the common hepatic artery (table 1). Instrumentation and data acquisition to record the normal values was completed within 1.5 h. Systemic and hepatic hemodynamic parameters remained within normal ranges during this procedure. This procedure is challenging but feasible. We have already applied this procedure to assess hepatic hemodynamics in normal mice as well as during 70% partial hepatectomy and in liver lobe clamping experiments. Mean PVP after resection (n= 20), was 11.41 ± 2.94 cmH2O which was significantly higher (P<0.05) than before resection (6.87 ± 2.39 cmH2O). The results of liver lobe clamping experiment indicated that this monitoring procedure is sensitive and suitable for detecting small changes in portal pressure and portal flow rate. In conclusion, this procedure is reliable in the hands of an experienced micro-surgeon but should be limited to experiments where mice are absolutely needed.


Subject(s)
Liver Circulation/physiology , Liver/blood supply , Animals , Blood Pressure/physiology , Computer Systems , Hemodynamics , Hepatectomy , Hepatic Artery/physiology , Mice , Monitoring, Physiologic/methods , Portal Vein/physiology
11.
Histochem Cell Biol ; 142(6): 667-76, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25059648

ABSTRACT

Liver dysfunction is a serious complication in the early phase following major liver resection or liver transplantation and might be aggravated by the translocation of bacteria and lipopolysaccharide (LPS). As a preventive strategy, granulocyte colony-stimulating factor (G-CSF) is prophylactically applied in patients who are subjected to major surgery. However, we previously demonstrated that G-CSF can induce LPS sensitization. In this study, we aimed to evaluate the effects of G-CSF pretreatment on hepatic microcirculatory disturbances and postoperative liver dysfunction after 70 % partial hepatectomy (PH) in rats. PH alone was well tolerated by all animals (100 % survival rate, slight liver damage and inflammation). LPS application after 70 % PH caused moderate inflammation, microcirculatory disturbances and hepatic damage and led to a 24-h survival rate of 30 % after the operations. In the G-CSF-LPS-PH group, all of the rats died within 4 h with severe inflammatory responses and liver damage (i.e., pronounced erythrocyte congestion and neutrophil infiltration). Portal hypertension and microcirculatory disorders (i.e., inhomogeneous perfusion, sinusoidal dilatation and reductions on functional capillary density) were more pronounced in the G-CSF-LPS-PH group. In conclusion, increased circulating LPS levels were associated with an imbalanced inflammatory response and microcirculatory dysfunction that preceded liver damage and subsequent dysfunction following surgery. G-CSF-pretreatment aggravated microcirculatory disturbances and liver damage, which might have been related to G-CSF-induced LPS sensitization.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hepatectomy , Lipopolysaccharides/toxicity , Liver Diseases/prevention & control , Liver/drug effects , Microcirculation/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Drug Interactions , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Liver/injuries , Liver/surgery , Male , Rats , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL
...