Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
2.
STAR Protoc ; 2(2): 100534, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34027491

ABSTRACT

Tumor organoids and patient-derived orthotopic xenografts (PDOXs) are some of the most valuable pre-clinical tools in cancer research. In this protocol, we describe efficient derivation of organoids and PDOX models from glioma patient tumors. We provide detailed steps for organoid culture, intracranial implantation, and detection of tumors in the brain. We further present technical adjustments for standardized functional assays and drug testing. For complete details on the use and execution of this protocol, please refer to Golebiewska et al. (2020).


Subject(s)
Brain Neoplasms/pathology , Drug Screening Assays, Antitumor/methods , Glioma/pathology , Heterografts , Organoids , Animals , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Female , Heterografts/cytology , Heterografts/drug effects , Humans , Male , Mice , Organoids/cytology , Organoids/drug effects , Tumor Cells, Cultured/cytology
3.
Nat Commun ; 12(1): 3275, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34045451

ABSTRACT

Despite advancements in human pluripotent stem cells (hPSCs) differentiation protocols to generate appropriate neuronal progenitors suitable for transplantation in Parkinson's disease, resultant grafts contain low proportions of dopamine neurons. Added to this is the tumorigenic risk associated with the potential presence of incompletely patterned, proliferative cells within grafts. Here, we utilised a hPSC line carrying a FailSafeTM suicide gene (thymidine kinase linked to cyclinD1) to selectively ablate proliferative cells in order to improve safety and purity of neural transplantation in a Parkinsonian model. The engineered FailSafeTM hPSCs demonstrated robust ventral midbrain specification in vitro, capable of forming neural grafts upon transplantation. Activation of the suicide gene within weeks after transplantation, by ganciclovir administration, resulted in significantly smaller grafts without affecting the total yield of dopamine neurons, their capacity to innervate the host brain or reverse motor deficits at six months in a rat Parkinsonian model. Within ganciclovir-treated grafts, other neuronal, glial and non-neural populations (including proliferative cells), were significantly reduced-cell types that may pose adverse or unknown influences on graft and host function. These findings demonstrate the capacity of a suicide gene-based system to improve both the standardisation and safety of hPSC-derived grafts in a rat model of Parkinsonism.


Subject(s)
Cell Engineering/methods , Genes, Transgenic, Suicide , Parkinson Disease, Secondary/therapy , Stem Cell Transplantation/methods , Animals , Apoptosis/genetics , Cell Differentiation , Cell Line , Cell Proliferation/genetics , Disease Models, Animal , Dopaminergic Neurons/physiology , Female , Genes, bcl-1/genetics , Heterografts/cytology , Heterografts/pathology , Human Embryonic Stem Cells/physiology , Humans , Male , Mesencephalon/cytology , Mesencephalon/pathology , Oxidopamine/administration & dosage , Oxidopamine/toxicity , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Rats , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/standards , Thymidine Kinase/genetics
4.
PLoS One ; 15(6): e0233899, 2020.
Article in English | MEDLINE | ID: mdl-32584883

ABSTRACT

Studies of the normal functions and diseases of the prostate request in vivo models that maintain the tissue architecture and the multiple-cell type compartments of human origin in order to recapitulate reliably the interactions of different cell types. Cell type-specific transcriptomes are critical to reveal the roles of each cell type in the functions and diseases of the prostate. A primary prostate tissue xenograft model was developed using fresh human prostate tissue specimens transplanted onto male mice that were castrated surgically and implanted with a device to maintain circulating testosterone levels comparable to adult human males. Endothelial cells and epithelial cells were isolated from 7 fresh human prostate tissue specimens and from primary tissue xenografts established from 9 fresh human prostate tissue specimens, using antibody-conjugated magnetic beads specific to human CD31 and human EpCAM, respectively. Transcriptomes of endothelial, epithelial and stromal cell fractions were obtained using RNA-Seq. Global and function-specific gene expression profiles were compared in inter-cell type and inter-tissue type manners. Gene expression profiles in the individual cell types isolated from xenografts were similar to those of cells isolated from fresh tissue, demonstrating the value of the primary tissue xenograft model for studies of the inter-relationships between prostatic cell types and the role of such inter-relationships in organ development, disease progression, and response to drug treatments.


Subject(s)
Endothelial Cells/metabolism , Heterografts/cytology , Prostate/cytology , Transcriptome , Animals , Endothelial Cells/cytology , Epithelial Cell Adhesion Molecule/metabolism , Humans , Male , Mice , Mice, Nude , Models, Animal , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
5.
Viruses ; 11(9)2019 08 29.
Article in English | MEDLINE | ID: mdl-31470671

ABSTRACT

Pigs are used as potential donor animals for xenotransplantation. However, porcine endogenous retrovirus (PERV), shown to infect both human and non-human primate (NHP) cells in vitro, presents a risk of transmission to humans in xenotransplantation. In this study, we analyzed PERV transmission in various organs after pig-to-NHP xenotransplantation. We utilized pig-to-NHP xenotransplant tissue samples obtained using two types of transgenic pigs from the National Institute of Animal Science (NIAS, Republic of Korea), and examined them for the existence of PERV genes in different organs via PCR and RT-PCR with specific primers. To determine PERV insertion into chromosomes, inverse PCR using PERV long terminal repeat (LTR) region-specific primers was conducted. The PERV gene was not detected in NHP organs in cardiac xenotransplantation but detected in NHP bladders in renal xenotransplantation. The insertion experiment confirmed that PERVs originate from porcine donor cells rather than integrated provirus in the NHP chromosome. We also demonstrate the presence of pig cells in the NHP bladder after renal xenotransplantation using specific-porcine mitochondrial DNA gene PCR. The PERV sequence was detected in the bladder of NHPs after renal xenotransplantation by porcine cell-microchimerism but did not integrate into the NHP chromosome.


Subject(s)
Endogenous Retroviruses/isolation & purification , Heterografts/virology , Kidney Transplantation/adverse effects , Transplantation, Heterologous/adverse effects , Urinary Bladder/virology , Animals , Animals, Genetically Modified , Chimerism , Cytochromes b/genetics , Endogenous Retroviruses/genetics , Genes, Viral/genetics , Heterografts/cytology , Humans , Macaca mulatta , Swine , Urinary Bladder/cytology
6.
Cell Death Dis ; 10(4): 299, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30931931

ABSTRACT

About 20% of prostate cancer (PCa) patients progress to metastatic disease. Metabolic syndrome (MeS) is a pathophysiological disorder that increases PCa risk and aggressiveness. C-terminal binding protein (CTBP1) is a transcriptional corepressor that is activated by high-fat diet (HFD). Previously, our group established a MeS/PCa mice model that identified CTBP1 as a novel link associating both diseases. Here, we integrated in vitro (prostate tumor cell lines) and in vivo (MeS/PCa NSG mice) models with molecular and cell biology techniques to investigate MeS/CTBP1 impact over PCa progression, particularly over cell adhesion, mRNA/miRNA expression and PCa spontaneous metastasis development. We found that CTBP1/MeS regulated expression of genes relevant to cell adhesion and PCa progression, such as cadherins, integrins, connexins, and miRNAs in PC3 xenografts. CTBP1 diminished PCa cell adhesion, membrane attachment to substrate and increased filopodia number by modulating gene expression to favor a mesenchymal phenotype. NSG mice fed with HFD and inoculated with CTBP1-depleted PC3 cells, showed a decreased number and size of lung metastases compared to control. Finally, CTBP1 and HFD reduce hsa-mir-30b-5p plasma levels in mice. This study uncovers for the first time the role of CTBP1/MeS in PCa progression and its molecular targets.


Subject(s)
Alcohol Oxidoreductases/metabolism , Cell Adhesion/genetics , DNA-Binding Proteins/metabolism , Metabolic Syndrome/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Alcohol Oxidoreductases/genetics , Animals , DNA-Binding Proteins/genetics , Diet, High-Fat , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Heterografts/cytology , Heterografts/metabolism , Humans , Male , Metabolic Syndrome/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Neoplasm Metastasis , PC-3 Cells , Prostatic Neoplasms/pathology , Pseudopodia/genetics , Pseudopodia/metabolism , RNA, Messenger/metabolism
7.
Int J Mol Sci ; 19(7)2018 Jul 15.
Article in English | MEDLINE | ID: mdl-30011964

ABSTRACT

Cultivation of autologous human tenocytes in a cell-free xenogenic extracellular tendon matrix (xECM) could present an approach for tendon reconstruction. The aim of this study was to achieve tendon-like tissue formation by implanting decellularized porcine Achilles tendons recellularized with human hamstring tendon-derived tenocytes into nude mice. The structure of decellularized xECM was histologically monitored before being dynamically reseeded with human tenocytes. After 6⁻12 weeks in vivo, construct quality was monitored using macroscopical and histological scoring systems, vitality assay and quantitative DNA and glycosaminoglycan (GAG) assays. For comparison to tendon xECM, a synthetic polyglycolic acid (PGA) polymer was implanted in a similar manner. Despite decellularized xECM lost some GAGs and structure, it could be recellularized in vitro with human tenocytes, but the cell distribution remained inhomogeneous, with accumulations at the margins of the constructs. In vivo, the xECM constructs revealed in contrast to the PGA no altered size, no inflammation and encapsulation and a more homogeneous cell distribution. xECM reseeded with tenocytes showed superior histological quality than cell-free implanted constructs and contained surviving human cells. Their DNA content after six and 12 weeks in vivo resembled that of native tendon and xECM recellularized in vitro. Results suggest that reseeded decellularized xECM formed a tendon-like tissue in vivo.


Subject(s)
Achilles Tendon/metabolism , Extracellular Matrix/transplantation , Heterografts/metabolism , Tenocytes/transplantation , Achilles Tendon/cytology , Animals , Cells, Cultured , Glycosaminoglycans/metabolism , Heterografts/cytology , Humans , Mice, Nude , Polyglycolic Acid/metabolism , Swine , Tendon Injuries/surgery , Tenocytes/cytology , Tissue Engineering/methods , Transplantation, Heterologous
8.
Plast Reconstr Surg ; 142(4): 927-938, 2018 10.
Article in English | MEDLINE | ID: mdl-29979369

ABSTRACT

BACKGROUND: Adipose-derived stem cells are used to enhance fat graft survival. However, their mechanism of action when grafted is controversial. The grafted cells can be replaced by connective tissue or survive at the recipient site and differentiate. Insulin is a powerful agent used to induce their differentiation to adipocytes. The purpose of this study was to elucidate the fate of grafted adipose-derived stem cells in nonvascularized fat grafts with or without insulin. METHODS: Fat was harvested from a female human donor who had undergone reduction mammaplasty. The authors also isolated and cultured adipose-derived stem cells expressing green fluorescent protein from transgenic Sprague-Dawley rats. Injection of free fat graft, the adipose-derived stem cells, phosphate-buffered saline, and insulin was performed on the four paravertebral points of the back of each mouse (n = 30) as follows: group A (control group) received adipose tissue and phosphate-buffered saline; group B received adipose tissue and adipose-derived stem cells; group C received adipose tissue, adipose-derived stem cells, and phosphate-buffered saline; and group D received adipose tissue, adipose-derived stem cells, and insulin. Green fluorescent protein expression was evaluated using an in vivo imaging system. The volume of transplanted fat was evaluated at 8 weeks after graft with six histologic parameters. The fat graft was immunostained with green fluorescent protein, 4',6-diamidino-2-phenylindole, and perilipin. Statistical analysis was performed using a one-way analysis of variance test. RESULTS: The fat graft volume was significantly higher in group D (p < 0.05). Histologic examination revealed reduced fibrosis and increased cysts, vacuoles, integrity, and vascularity in group D. The green fluorescent protein and perilipin co-positive area was more apparent in group D compared with groups B and C. CONCLUSION: Insulin could enhance the survival and differentiation of adipose-derived stem cells in nonvascularized fat grafts.


Subject(s)
Adipose Tissue/transplantation , Cell Differentiation/drug effects , Insulin/pharmacology , Adipocytes/drug effects , Animals , Female , Heterografts/cytology , Heterografts/drug effects , Humans , Male , Mice, Nude , Microscopy, Confocal , Microscopy, Fluorescence , Middle Aged , Rats, Sprague-Dawley , Stem Cell Transplantation/methods , Stem Cells/drug effects , Transplantation, Heterologous/methods
9.
Transplantation ; 102(10): e413-e423, 2018 10.
Article in English | MEDLINE | ID: mdl-29975241

ABSTRACT

BACKGROUND: There is a strong rationale to pursue the use of neonatal porcine islets (NPIs) as an unlimited source of islets for clinical xenotransplantation. Because NPIs are composed of immature insulin producing beta (ß) cells and ductal precursor cells, they provide an ideal model to examine culture conditions to enhance ß cell proliferation and/or ß cell neoformation from ductal cells. In an attempt to optimize the potential of NPIs as a source of ß cell grafts, we used an in vitro differentiation protocol and measured its effect on the functional maturation and differentiation of NPIs. METHODS: Pancreata from 1- to 3-day-old neonatal pigs were digested and cultured in standard Ham's F10 media for 5 days. Each independent preparation was then further cultured in Dulbecco's modified Eagle medium nutrient mixture-F12 differentiation media containing growth factors added in a stepwise fashion, or cultured in control Ham's F10 media. After 20 days in culture, islets were assessed for insulin secretory capacity, cellular composition, gene expression, and metabolic activity after transplantation in immunodeficient mice with diabetes. RESULTS: Compared with control islets, differentiated islets exhibited a significantly higher proportion of endocrine cells, proliferating cell nuclear antigen double positive ß cells, and an enhanced glucose-stimulated insulin secretory activity. Mice transplanted with differentiated islets had significantly lower blood glucose values at weeks 18 and 20 compared with nondifferentiated controls and were shown to be more glucose tolerant. CONCLUSIONS: Culturing NPIs in a 20-day stepwise differentiation media increases the proportion of endocrine cells and augments both in vitro and in vivo function of the islets.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/methods , Islets of Langerhans/physiology , Tissue Culture Techniques/methods , Transplantation, Heterologous/methods , Animals , Animals, Newborn , Blood Glucose , Cell Differentiation , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Heterografts/cytology , Heterografts/physiology , Humans , Insulin/metabolism , Islets of Langerhans/cytology , Male , Mice , Streptozocin/toxicity , Sus scrofa , Treatment Outcome
10.
J Mol Neurosci ; 65(1): 60-73, 2018 May.
Article in English | MEDLINE | ID: mdl-29705933

ABSTRACT

Mesenchymal stem cells (MSCs) are regarded as an immune privileged cell type with numerous regeneration-promoting effects. The in vivo behavior of MSC and underlying mechanisms leading to their regenerative effects are largely unknown. The aims of this study were to comparatively investigate the in vivo behavior of canine (cMSC), human (hMSC), and murine MSC (mMSC) following intra-cerebroventricular transplantation. At 7 days post transplantation (dpt), clusters of cMSC, hMSC, and mMSC were detected within the ventricular system. At 49 dpt, cMSC-transplanted mice showed clusters mostly consisting of extracellular matrix lacking transplanted MSC. Similarly, hMSC-transplanted mice lacked MSC clusters at 49 dpt. Xenogeneic MSC transplantation was associated with a local T lymphocyte-dominated immune reaction at both time points. Interestingly, no associated inflammation was observed following syngeneic mMSC transplantation. In conclusion, transplanted MSC formed intraventricular cell clusters and exhibited a short life span in vivo. Xenogeneically in contrast to syngeneically transplanted MSC triggered a T cell-mediated graft rejection indicating that MSCs are not as immune privileged as previously assumed. However, MSC may mediate their effects by a "hit and run" mechanism and future studies will show whether syngeneically or xenogeneically transplanted MSCs exert better therapeutic effects in animals with CNS disease.


Subject(s)
Cerebral Ventricles/surgery , Heterografts/cytology , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Cerebral Ventricles/cytology , Dogs , Female , Graft Rejection/immunology , Heterografts/immunology , Humans , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
11.
Tissue Eng Part C Methods ; 24(5): 280-288, 2018 05.
Article in English | MEDLINE | ID: mdl-29478384

ABSTRACT

Tissue antigenicity represents the main limitation for the use of xenografts in clinical practice. To eliminate xenoantigens and avoid graft rejection in human, decellularization is often used to remove all immunoreactive components from the extracellular matrix (ECM). After decellularization, acellular scaffolds are required to be investigated regarding the presence of antigens, but commonly used detection methods solely focus on known xenoantigens such as alpha Gal (Galα1,3-Galß1-4GlcNAc-R) or major histocompatibility complex-I (MHC-I). However, there are unknown xenoantigens that escape the standard methods. To evaluate the immunological potential of xenogenic tissues, new in vitro methods need to be developed. Therefore, we established a novel human serum-based approach, including dot blot, Western blot, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA). With these methods, we analyzed protein extracts and tissue samples of native and decellularized bovine carotid arteries. All methods verified an effective removal of potential immunogens from the ECM through decellularization, and relative quantification with ELISA showed that 99.9% (p < 0.01) of antigenic components were successfully eliminated. We compared our human serum-based methods with commonly used assays for the detection of alpha Gal and MHC-I. Our results showed highly increased sensitivity for xenoantigens using the human serum antibody pool. This novel in vitro detection system allows the direct determination of the immunogenic potential of xenografts and is a vast improvement in comparison to the methods used so far. That way, it is possible to optimize the decellularization process to prevent hyperacute graft rejection in patients.


Subject(s)
Heterografts/immunology , Animals , Carotid Arteries/cytology , Cattle , Cells, Cultured , Endothelial Cells/cytology , Heterografts/cytology , Humans , Reference Standards , Serum/metabolism
12.
Xenotransplantation ; 24(4)2017 07.
Article in English | MEDLINE | ID: mdl-28741700

ABSTRACT

When clinical xenotransplantation is introduced, the costs associated with acquisition of a genetically engineered pig organ are as yet unknown. How will these costs compare with those currently associated with the acquisition of deceased human organs? An understanding of the financial aspects of deceased organ and tissue procurement in the USA is therefore worthwhile. We have therefore attempted to review certain economic aspects of non-profit and for-profit organizations that provide cadaveric organs and/or tissues for purposes of transplantation into patients with end-stage organ failure, cellular deficiencies, or in need of reconstructive procedures. We briefly consider the laws, organizations, and business practices that govern the acquisition, processing, and/or distribution of cadaveric organs and tissues, and the economic implications of industry practices. In particular, we explore and highlight what we perceive as a lack of transparency and oversight with regard to financial practices, and we question whether donor families would be entirely happy with the business environment that has developed from their altruistic donations. Until xenotransplantation becomes established clinically, which will negate the need for any system of organ procurement and allocation, we suggest that those involved in organ and cell transplantation, as well as those who participate in reconstructive surgery, should take responsibility to ensure that the financial practices associated with procurement are transparent, and overseen/regulated by a responsible authority. We suggest the major transplant societies should take a lead in this respect. The ability to acquire a genetically engineered pig organ whenever required through a simple commercial transaction (as in the acquisition of a life-saving drug) will be greatly to the patient's benefit.


Subject(s)
Heterografts/cytology , Tissue and Organ Procurement , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Humans , Swine , Tissue Donors , Transplantation, Heterologous/methods , United States
13.
Xenotransplantation ; 24(4)2017 07.
Article in English | MEDLINE | ID: mdl-28623861

ABSTRACT

BACKGROUND: Recently, significant progress in both safety and efficacy has been achieved in the field of xenotransplantation, as exemplified by results from the first clinical trials of porcine islet transplantation. It would be of interest to learn whether the attitude of the clinical staff involved in such trials changes as the trials are carried out in their own hospital. METHODS: One hundred and four clinical staff members from the Eva Peron Hospital of San Martin (Buenos Aires, Argentina) where clinical trials of islet xenotransplantation have been performed and 92 similar staff members from the Diego Thompson Hospital (Buenos Aires, Argentina) where no such xenotransplantation has been carried out participated in the study. Data were collected anonymously using questionnaires. RESULTS: Statistically significant differences between the acceptance of xenotransplantation by clinical personnel in a hospital that had carried out clinical xenotransplantation trials were observed when compared with the acceptance of a similar staff from the hospital that had not carried out such trials. CONCLUSION: This study shows that involvement in clinical xenotransplantation trials significantly changes the attitude of the clinical staff towards this technology and suggests that better information given to the society may increase acceptance of the xenotransplantation.


Subject(s)
Heterografts/cytology , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Kidney Transplantation , Transplantation, Heterologous , Animals , Diabetes Mellitus, Type 1 , Heterografts/immunology , Humans , Islets of Langerhans Transplantation/methods , Kidney/cytology , Kidney Transplantation/methods , Patient Selection , Surveys and Questionnaires , Swine , Transplantation, Heterologous/methods
14.
Mol Hum Reprod ; 23(3): 141-154, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28130393

ABSTRACT

Study question: Does irradiation evoke adverse effects in germ and somatic cells in testis xenografts from prepubertal monkeys? Summary answer: In addition to the expected depletion of germ cells, a dose-dependent effect of irradiation was observed at the mRNA and protein level in Sertoli and peritubular myoid cells. What is known already: Testicular irradiation studies in monkeys have focused on the dose-dependent effects on germ cells. Previous studies using intact animals or xenografts reported that germ cells are highly sensitive to irradiation. Their depletion was demonstrated by morphometric and histological analyses. The effect of irradiation on expression of Sertoli and peritubular myoid cell markers, however, has not yet been described. Study design, size, duration: The testes of two prepubertal macaques (Macaca fascicularis) were dissected into testicular fragments. Fragments were randomly exposed in vitro to one of the following three doses of irradiation: 0 Gy, n = 60; 1 Gy, n = 54; 4 Gy, n = 72. Non-irradiated control fragments (0 Gy) were placed into the Faxitron for 6.6 min without irradiation. For 1 Gy and 4 Gy irradiation was applied for 1.7 and 6.6 min, respectively. Grafts were then either immediately analyzed or subcutaneously implanted under the back skin of 39 nude mice and analyzed after 6.5 months. Participants/materials setting methods: Post grafting, 133 testicular xenografts were retrieved. The body weight, serum testosterone level and seminal vesical weight of the host mice as well as the number and weight of retrieved grafts were determined. Larger grafts were used to evaluate both mRNA expression profiles and protein expression patterns. In total, 71 testicular fragments were used for morphometric and histological analysis while 68 fragments were analyzed for gene expression. For PCR arrays, M. fascicularis-specific primer sequences were employed. Irradiation-induced changes in the transcript levels of 34 marker genes were determined for each testicular graft. The effects of irradiation on peritubular myoid cells and Sertoli cells were confirmed by immunohistochemical analysis of chemokine (C-X-C motif) ligand type 11 (CXCL11), alpha smooth muscle actin (SMA) and chemokine (C-X-C motif) ligand type 12 (CXCL12). Main results and the role of chance: The four testes gave rise to 106 xenografts, which were individually analyzed, limiting the role of chance despite using only two monkeys in the study. Prior to grafting, the two donors displayed spermatogonia as the most advanced germ cell type in 95% and 70% of seminiferous tubules, respectively, while remaining tubules contained SCO. No spermatocytes were encountered prior to grafting in either monkey. After 6.5 months, non-irradiated grafts displayed spermatocytes in 15.4% and 1.8% of seminiferous tubules indicating an induction of meiosis. Irradiation resulted in a complete absence of spermatocytes. The percentage of seminiferous tubules containing spermatogonia declined in a dose-dependent manner. In non-irradiated xenografts, ~40% of tubules contained spermatogonia. This proportion was reduced to 3.4% and 4.3% in the 1 Gy treated group and to 1.3% and 0.2% in 4 Gy irradiated grafts. A dose-dependent decline in mRNA levels of selected germ cell marker genes supported the morphologically detected loss of germ cells. Irradiation had no effect on CXCL12 transcript levels. At the protein level, CXCL12-positive Sertoli cells were most abundant in the 1 Gy group compared to the 4 Gy group (P < 0.05), indicating a potential role of CXCL12 during recovery of primate spermatogenesis. The most prominent radiation-evoked changes were for CXCL11, which was localized to smooth muscle cells of blood vessels and seminiferous tubules. Transcript levels declined in a dose-dependent manner in grafts from both monkeys (MM687: P < 0.01 (0 Gy versus 4 Gy), MM627: P < 0.05 (0 Gy versus 4 Gy), P < 0.001 (1 Gy versus 4 Gy)). CXCL11 patterns of protein expression revealed irradiation-dependent changes as well. That peritubular cells are affected by X-irradiation was substantiated by changes at the transcript level between 1 and 4 Gy exposed groups (P < 0.01) and at the protein level of SMA (P < 0.05, 0 Gy versus 4 Gy). Large scale data: n/a. Limitations, reasons for caution: The spermatogonial stem cell system in primates is remarkably different from rodents. Therefore, data from a non-human primate may be more relevant to man. However, species-specific differences amongst primates cannot be fully excluded and the use of only two donors may raise concerns toward the generalization of the findings. There may also be important differences across the prepubertal period (e.g. infancy, early childhood) that are not represented by the ages included in the present study. Wider implications of the findings: This study is the first to indicate relevant testicular somatic cell responses following irradiation of prepubertal primate tissue. In addition to the well-known depletion of germ cells, the changes in Sertoli, and in particular peritubular myoid, cells may have important consequences for spermatogenic recovery. These novel findings should be taken into consideration when irradiation effects are assessed in tumor survivors. Study funding and competing interest(s): Interdisciplinary Center for Clinical Research (IZKF) Münster (Schl2/001/13) and the Excellence Cluster 'Cells in Motion' at the University Münster. There are no conflicts of interest to declare.


Subject(s)
Choristoma , Heterografts/radiation effects , Seminiferous Tubules/radiation effects , Sertoli Cells/radiation effects , Spermatogenesis/radiation effects , Spermatogonia/radiation effects , Actins/genetics , Actins/metabolism , Animals , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Gamma Rays , Gene Expression Regulation , Heterografts/cytology , Heterografts/metabolism , Macaca fascicularis , Male , Mice , Mice, Nude , Seminiferous Tubules/cytology , Seminiferous Tubules/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Sexual Maturation/physiology , Signal Transduction , Skin , Spermatogonia/cytology , Spermatogonia/metabolism , Transplantation, Heterologous
15.
Arthroscopy ; 33(2): 374-386, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27692557

ABSTRACT

PURPOSE: To evaluate the biological, immunological, and biomechanical properties of a scaffold derived by architectural modification of a fresh-frozen porcine patella tendon using a decellularization protocol that combines physical, chemical, and enzymatic modalities. METHODS: Porcine patellar tendons were processed using a decellularization and oxidation protocol that combines physical, chemical, and enzymatic modalities. Scaffolds (n = 88) were compared with native tendons (n = 70) using histologic, structural (scanning electron microscopy, porosimetry, and tensile testing), biochemical (mass spectrometry, peracetic acid reduction, DNA quantification, alpha-galactosidase [α-gal] content), as well as in vitro immunologic (cytocompatibility, cytokine induction) and in vivo immunologic nonhuman primate analyses. RESULTS: A decrease in cellularity based on histology and a significant decrease in DNA content were observed in the scaffolds compared with the native tendon (P < .001). Porosity and pore size were increased significantly (P < .001). Scaffolds were cytocompatible in vitro. There was no difference between native tendons and scaffolds when comparing ultimate tensile load, stiffness, and elastic modulus. The α-gal xenoantigen level was significantly lower in the decellularized scaffold group compared with fresh-frozen, nondecellularized tissue (P < .001). The in vivo immunological response to implanted scaffolds measured by tumor necrosis factor-α and interleukin-6 levels was significantly (P < .001) reduced compared with untreated controls in vitro. These results were confirmed by an attenuated response to scaffolds in vivo after implantation in a nonhuman primate model. CONCLUSIONS: Porcine tendon was processed via a method of decellularization and oxidation to produce a scaffold that possessed significantly less inflammatory potential than a native tendon, was biocompatible in vitro, of increased porosity, and with significantly reduced amounts of α-gal epitope while retaining tensile properties. CLINICAL RELEVANCE: Porcine-derived scaffolds may provide a readily available source of material for musculoskeletal reconstruction and repair while eliminating concerns regarding disease transmission and the morbidity of autologous harvest.


Subject(s)
Heterografts/cytology , Tendons/transplantation , Tissue Scaffolds , Animals , Ligaments/cytology , Ligaments/transplantation , Oxidation-Reduction , Swine , Tendons/cytology , Tendons/metabolism , Tensile Strength , alpha-Galactosidase/metabolism
16.
Xenotransplantation ; 23(6): 429-439, 2016 11.
Article in English | MEDLINE | ID: mdl-27678013

ABSTRACT

BACKGROUND: Increased abdominal fat and chronic inflammation in the expanded adipose tissue of obesity contribute to the development of insulin resistance and type 2 diabetes mellitus (T2D). The emerging immunoregulatory and anti-inflammatory properties of Sertoli cells have prompted their application to experimental models of autoimmune/inflammatory disorders, including diabetes. The main goal of this work was to verify whether transplantation of microencapsulated prepubertal porcine Sertoli cells (MC-SC) in the subcutaneous abdominal fat depot of spontaneously diabetic and obese db/db mice (homozygous for the diabetes spontaneous mutation [Leprdb ]) would: (i) improve glucose homeostasis and (ii) modulate local and systemic immune response and adipokines profiles. METHODS: Porcine prepubertal Sertoli cells were isolated, according to previously established methods and enveloped in Barium alginate microcapsules by a mono air-jet device. MC-SC were then injected in the subcutaneous abdominal fat depot of db/db mice. RESULTS: We have preliminarily shown that graft of MC-SC restored glucose homeostasis, with normalization of glycated hemoglobin values with improvement of the intraperitoneal glucose tolerance test in 60% of the treated animals. These results were associated with consistent increase, in the adipose tissue, of uncoupling protein 1 expression, regulatory B cells, anti-inflammatory macrophages and a concomitant decrease of proinflammatory macrophages. Furthermore, the treated animals showed a reduction in inducible NOS and proinflammatory molecules and a significant increase in an anti-inflammatory cytokine such as IL-10 along with concomitant rise of circulating adiponectin levels. The anti-hyperglycemic graft effects also emerged from an increased expression of GLUT-4, in conjunction with downregulation of GLUT-2, in skeletal muscle and liver, respectively. CONCLUSIONS: Preliminarily, xenograft of MC-SC holds promises for an effective cell therapy approach for treatment of experimental T2D.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/immunology , Heterografts/cytology , Homeostasis/immunology , Sertoli Cells/transplantation , Transplantation, Heterologous , Adipose Tissue/cytology , Animals , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/therapy , Drug Compounding , Glucose Tolerance Test/methods , Heterografts/immunology , Insulin Resistance/physiology , Male , Mice, Transgenic , Swine , Transplantation, Heterologous/methods
17.
Gen Comp Endocrinol ; 230-231: 48-56, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26972155

ABSTRACT

Fish germ cell transplantation presents several important potential applications for aquaculture, including the preservation of germplasm from endangered fish species with high genetic and commercial values. Using this technique in studies developed in our laboratory with adult male Nile tilapias (Oreochromis niloticus), all the necessary procedures were successfully established, allowing the production of functional sperm and healthy progeny approximately 2months after allogeneic transplantation. In the present study, we evaluated the viability of the adult Nile tilapia testis to generate sperm after xenogeneic transplant of germ cells from sexually mature Jundia catfish (Rhamdia quelen) that belong to a different taxonomic order. Therefore, in order to investigate at different time-periods post-transplantation, the presence and development of donor PKH26 labeled catfish germ cells were followed in the tilapia seminiferous tubules. From 7 to 20days post-transplantation, only PKH26 labeled spermatogonia were observed, whereas spermatocytes at different stages of development were found at 70days. Germ cell transplantation success and progression of spermatogenesis were indicated by the presence of labeled PKH26 spermatids and sperm on days 90 and 120 post-transplantation, respectively. Confirming the presence of the catfish genetic material in the tilapia testis, all recipient tilapias evaluated (n=8) showed the genetic markers evaluated. Therefore, we demonstrated for the first time that the adult Nile tilapia testis offers the functional conditions for development of spermatogenesis with sperm production from a fish species belonging to a different order, which provides an important new venue for aquaculture advancement.


Subject(s)
Catfishes/metabolism , Cell Transplantation , Heterografts/cytology , Spermatozoa/cytology , Testis/cytology , Tilapia/metabolism , Transplantation, Heterologous , Animals , Aquaculture/methods , Catfishes/genetics , Conservation of Natural Resources/methods , Endangered Species , Heterografts/growth & development , Male , Seminiferous Tubules/cytology , Spermatids/cytology , Spermatids/growth & development , Spermatids/metabolism , Spermatocytes/cytology , Spermatocytes/growth & development , Spermatocytes/metabolism , Spermatogenesis , Spermatogonia/cytology , Spermatogonia/growth & development , Spermatogonia/metabolism , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/physiology , Tilapia/genetics
18.
Diabetes ; 65(5): 1297-309, 2016 05.
Article in English | MEDLINE | ID: mdl-26740603

ABSTRACT

Pancreatic progenitors derived from human embryonic stem cells (hESCs) are a potential source of transplantable cells for treating diabetes and are currently being tested in clinical trials. Yet, how the milieu of pancreatic progenitor cells, including exposure to different factors after transplant, may influence their maturation remains unclear. Here, we examined the effect of thyroid dysregulation on the development of hESC-derived progenitor cells in vivo. Hypothyroidism was generated in SCID-beige mice using an iodine-deficient diet containing 0.15% propyl-2-thiouracil, and hyperthyroidism was generated by addition of L-thyroxine (T4) to drinking water. All mice received macroencapsulated hESC-derived progenitor cells, and thyroid dysfunction was maintained for the duration of the study ("chronic") or for 4 weeks posttransplant ("acute"). Acute hyperthyroidism did not affect graft function, but acute hypothyroidism transiently impaired human C-peptide secretion at 16 weeks posttransplant. Chronic hypothyroidism resulted in severely blunted basal human C-peptide secretion, impaired glucose-stimulated insulin secretion, and elevated plasma glucagon levels. Grafts from chronic hypothyroid mice contained fewer ß-cells, heterogenous MAFA expression, and increased glucagon(+) and ghrelin(+) cells compared to grafts from euthyroid mice. Taken together, these data suggest that long-term thyroid hormone deficiency may drive the differentiation of human pancreatic progenitor cells toward α- and ε-cell lineages at the expense of ß-cell formation.


Subject(s)
Cell Differentiation , Diabetes Mellitus, Type 1/surgery , Disease Models, Animal , Heterografts/pathology , Human Embryonic Stem Cells/transplantation , Hypothyroidism/complications , Insulin-Secreting Cells/transplantation , Animals , Antithyroid Agents/poisoning , Biomarkers/blood , Biomarkers/metabolism , Cell Line , Cells, Immobilized/cytology , Cells, Immobilized/pathology , Cells, Immobilized/transplantation , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Heterografts/cytology , Heterografts/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Hyperthyroidism/chemically induced , Hyperthyroidism/complications , Hypothyroidism/etiology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Iodine/deficiency , Male , Mice, SCID , Propylthiouracil/poisoning , Random Allocation , Thyroxine/poisoning , Transplantation, Heterologous , Transplantation, Heterotopic
19.
Stem Cells Dev ; 25(4): 360-5, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26650921

ABSTRACT

Zebrafish is gaining increased popularity as a model organism to study stem cell biology. It also is widely used as model system to visualize human leukemic stem cells. However, xenotransplantation of primary human stem/progenitor cells has not been described. Here, we use casper pigmentation mutant fish that are transparent crossed to fli-GFP transgenic fish as recipients of red labeled human CD34(+) cells. We have investigated various conditions and protocols with the aim to monitor and visualize the fate of transplanted human CD34(+) cells. We here report successful use of casper mutant zebrafish embryos for the direct monitoring of human hematopoietic stem cell transplantation, differentiation, and trafficking in vivo.


Subject(s)
Cell Differentiation , Cell Movement , Cell Tracking/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Heterografts/cytology , Zebrafish/metabolism , Animals , Cell Lineage , Embryo, Nonmammalian/cytology , Humans , Models, Biological , Stromal Cells/metabolism
20.
Lab Invest ; 95(2): 207-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25485535

ABSTRACT

Many commercially available cell lines have been in culture for ages, acquiring phenotypes that differ from the original cancers from which these cell lines were derived. Therefore, research on new cell lines could improve the success rates of translational research in cancer. We have developed methods for the isolation and culture of human pancreatic ductal adenocarcinoma (PDAC) cells from murine xenografts of human PDAC. We hypothesize that phenotypes of PDAC cells are modified by in vitro culture conditions over time and by in vivo implantation. Patient-derived xenografts were created in immunodeficient mice using surgically resected tumor specimens. These murine xenografts were then used to establish human PDAC cell lines in culture. Earlier (<5) passage and later (>20) passage cell lines were evaluated separately regarding proliferation, cell cycle, genetic mutations, invasiveness, chemosensitivity, tumorigenesis, epithelial-mesenchymal transition (EMT) status, and proteomics. Later passage cells accelerated their doubling time and colony formation, and were more concentrated in the G0/G1 phase and less in the G2/M checkpoint phase. Later passage cells were more sensitive to gemcitabine and 5-fluorouracil than earlier passage cells, but all four new cell lines were more chemo-resistant compared with commercial ATCC cell lines. EMT induction was observed when establishing and passaging cell lines in vitro and furthermore by growing them as subcutaneous tumors in vivo. This study demonstrates a novel approach to the establishment of PDAC cell lines and observes a process by which newly established cell lines undergo phenotypic changes during in vitro culture and in vivo tumorigenesis. This may help explain differences of treatment effects often observed between experiments conducted in vitro, in vivo, and in human clinical trials.


Subject(s)
Cell Culture Techniques/methods , Epithelial-Mesenchymal Transition/physiology , Pancreatic Neoplasms/physiopathology , Phenotype , Animals , Blotting, Western , Cell Cycle/physiology , Cell Proliferation/physiology , Colony-Forming Units Assay , Deoxycytidine/analogs & derivatives , Fluorouracil , Heterografts/cytology , Heterografts/physiology , Humans , Immunohistochemistry , Mice , Neoplasm Invasiveness/physiopathology , Protein Array Analysis , Proteomics/methods , Tumor Cells, Cultured , Gemcitabine , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL