Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 16(756): eadi9548, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39018368

ABSTRACT

Immune rejection remains the major obstacle to long-term survival of allogeneic lung transplants. The expression of major histocompatibility complex molecules and minor histocompatibility antigens triggers allogeneic immune responses that can lead to allograft rejection. Transplant outcomes therefore depend on long-term immunosuppression, which is associated with severe side effects. To address this problem, we investigated the effect of genetically engineered transplants with permanently down-regulated swine leukocyte antigen (SLA) expression to prevent rejection in a porcine allogeneic lung transplantation (LTx) model. Minipig donor lungs with unmodified SLA expression (control group, n = 7) or with modified SLA expression (treatment group, n = 7) were used to evaluate the effects of SLA knockdown on allograft survival and on the nature and strength of immune responses after terminating an initial 4-week period of immunosuppression after LTx. Genetic engineering to down-regulate SLA expression was achieved during ex vivo lung perfusion by lentiviral transduction of short hairpin RNAs targeting mRNAs encoding ß2-microglobulin and class II transactivator. Whereas all grafts in the control group were rejected within 3 months, five of seven animals in the treatment group maintained graft survival without immunosuppression during the 2-year monitoring period. Compared with controls, SLA-silenced lung recipients had lower donor-specific antibodies and proinflammatory cytokine concentrations in the serum. Together, these data demonstrate a survival benefit of SLA-down-regulated lung transplants in the absence of immunosuppression.


Subject(s)
Gene Knockdown Techniques , Graft Survival , Histocompatibility Antigens Class I , Immunosuppression Therapy , Lung Transplantation , Animals , Swine , Graft Survival/immunology , Histocompatibility Antigens Class I/metabolism , Graft Rejection/immunology , Swine, Miniature , Histocompatibility Antigens Class II/metabolism , Transplantation, Homologous , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Lung/metabolism , Nuclear Proteins , Trans-Activators
2.
Hum Gene Ther ; 33(7-8): 460-471, 2022 04.
Article in English | MEDLINE | ID: mdl-34779223

ABSTRACT

Genetic engineering is a promising tool to repair genetic disorders, improve graft function, or reduce immune responses toward allografts. Ex vivo organ perfusion systems have the potential to mitigate ischemic-reperfusion injury, prolong preservation time, or even rescue organ function. We aim at combining both technologies to develop a modular platform allowing the genetic modification of vascularized composite (VC) allografts. Rat hind limbs were perfused ex vivo under subnormothermic conditions with lentiviral vectors. Specific perfusion conditions such as controlled pressure, temperature, and flow rates were optimized to support the genetic modification of the limbs. Genetic modification was detected in vascular, muscular, and dermal limb tissues. Remarkably, skin follicular and interfollicular keratinocytes, as well as endothelial cells showed stable transgene expression. Furthermore, levels of injury markers such as lactate, myoglobin, and lactate dehydrogenase, as well as histological analyses showed that ex vivo limb perfusion with lentiviral vectors did not cause tissue damage and limb cytokine secretion signatures were not significantly affected. The use of ex vivo VC perfusion in combination with lentiviral vectors allows an efficient and stable genetic modification representing a robust platform to genetically engineer limbs toward increasing graft survival after transplantation.


Subject(s)
Endothelial Cells , Graft Survival , Animals , Extremities , Perfusion , Rats , Temperature
3.
J Cell Mol Med ; 24(9): 5070-5081, 2020 05.
Article in English | MEDLINE | ID: mdl-32212307

ABSTRACT

Xenotransplantation of pancreatic islets offers a promising alternative to overcome the shortage of allogeneic donors. Despite significant advances, either immune rejection or oxygen supply in immune protected encapsulated islets remains major bottlenecks for clinical application. To decrease xenogeneic immune responses, we generated tissue engineered swine leucocyte antigen (SLA)-silenced islet cell clusters (ICC). Single-cell suspensions from pancreatic islets were generated by enzymatic digestion of porcine ICCs. Cells were silenced for SLA class I and class II by lentiviral vectors encoding for short hairpin RNAs targeting beta2-microglobulin or class II transactivator, respectively. SLA-silenced ICCs-derived cells were then used to form new ICCs in stirred bioreactors in the presence of collagen VI. SLA class I silencing was designed to reach a level of up to 89% and class II by up to 81% on ICCs-derived cells. Xenogeneic T cell immune responses, NK cell and antibody-mediated cellular-dependent immune responses were significantly decreased in SLA-silenced cells. In stirred bioreactors, tissue engineered islets showed the typical 3D structure and insulin production. These data show the feasibility to generate low immunogenic porcine ICCs after single-cell engineering and post-transduction islet reassembling that might serve as an alternative to allogeneic pancreatic islet cell transplantation.


Subject(s)
Histocompatibility Antigens Class I/immunology , Islets of Langerhans Transplantation/methods , Islets of Langerhans/metabolism , Animals , Antibodies/chemistry , Antibody Formation , Cell Survival , Cells, Cultured , Gene Silencing , Genetic Engineering/methods , Immunity, Cellular , Insulin/metabolism , Killer Cells, Natural/metabolism , Neoplasm Transplantation , Pancreas/metabolism , RNA Interference , Swine , T-Lymphocytes/metabolism , Transcriptional Activation , Transplantation, Heterologous
4.
Front Immunol ; 11: 265, 2020.
Article in English | MEDLINE | ID: mdl-32140158

ABSTRACT

Organ gene therapy represents a promising tool to correct diseases or improve graft survival after transplantation. Polymorphic variation of the major histocompatibility complex (MHC) antigens remains a major obstacle to long-term graft survival after transplantation. Previously, we demonstrated that MHC-silenced cells are protected against allogeneic immune responses. We also showed the feasibility to silence MHC in the lung. Here, we aimed at the genetic engineering of the kidney toward permanent silencing of MHC antigens in a rat model. We constructed a sub-normothermic ex vivo perfusion system to deliver lentiviral vectors encoding shRNAs targeting ß2-microglobulin and the class II transactivator to the kidney. In addition, the vector contained the sequence for a secreted nanoluciferase. After kidney transplantation (ktx), we detected bioluminescence in the plasma and urine of recipients of an engineered kidney during the 6 weeks of post-transplant monitoring, indicating a stable transgene expression. Remarkably, transcript levels of ß2-microglobulin and the class II transactivator were decreased by 70% in kidneys expressing specific shRNAs. Kidney genetic modification did not cause additional cell death compared to control kidneys after machine perfusion. Nevertheless, cytokine secretion signatures were altered during perfusion with lentiviral vectors as revealed by an increase in the secretion of IL-10, MIP-1α, MIP-2, IP-10, and EGF and a decrease in the levels of IL-12, IL-17, MCP-1, and IFN-γ. Biodistribution assays indicate that the localization of the vector was restricted to the graft. This study shows the potential to generate immunologically invisible kidneys showing great promise to support graft survival after transplantation and may contribute to reduce the burden of immunosuppression.


Subject(s)
Genetic Engineering/methods , Graft Survival , Kidney Transplantation/methods , Kidney/metabolism , Major Histocompatibility Complex/physiology , Nuclear Proteins/genetics , Trans-Activators/genetics , beta 2-Microglobulin/genetics , Animals , Cytokines/biosynthesis , Genetic Therapy , Male , Perfusion , Rats , Rats, Inbred Lew
5.
Sci Rep ; 9(1): 11318, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31383930

ABSTRACT

Low haemocompatibility of left ventricular assist devices (LVAD) surfaces necessitates anticoagulative therapy. Endothelial cell (EC) seeding can support haemocompatibility, however, the availability of autologous ECs is limited. In contrast, allogeneic ECs are readily available in sufficient quantity, but HLA disparities induce harmful immune responses causing EC loss. In this study, we investigated the feasibility of using allogeneic low immunogenic ECs to endothelialize LVAD sintered inflow cannulas (SIC). To reduce the immunogenicity of ECs, we applied an inducible lentiviral vector to deliver short-hairpins RNA to silence HLA class I expression. HLA class I expression on ECs was conditionally silenced by up to 70%. Sufficient and comparable endothelialization rates were achieved with HLA-expressing or HLA-silenced ECs. Cell proliferation was not impaired by cell-to-Sintered Inflow Cannulas (SIC) contact or by silencing HLA expression. The levels of endothelial phenotypic and thrombogenic markers or cytokine secretion profiles remained unaffected. HLA-silenced ECs-coated SIC exhibited reduced thrombogenicity. In contrast to native ECs, HLA-silenced ECs showed lower cell lysis rates when exposed to allogeneic T cells or specific anti-HLA antibodies. Allogeneic HLA-silenced ECs could potentially become a valuable source for LVAD endothelialization to reduce immunogenicity and correspondingly the need for anticoagulative therapy which can entail severe side effects.


Subject(s)
Allogeneic Cells/immunology , Bioprosthesis , Endothelial Cells/immunology , Heart-Assist Devices , Allogeneic Cells/cytology , Allogeneic Cells/metabolism , Bioprosthesis/adverse effects , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genes, MHC Class I , Heart-Assist Devices/adverse effects , Humans , Materials Testing , RNA Interference , RNA, Small Interfering/genetics , Thrombosis/etiology
6.
J Cell Mol Med ; 23(8): 5705-5714, 2019 08.
Article in English | MEDLINE | ID: mdl-31180181

ABSTRACT

In contrast to the whole liver, primary hepatocytes are highly immunogenic. Thus, alternative strategies of immunomodulation after hepatocyte transplantation are of special interest. Silencing of HLA class I expression is expected to reduce the strength of allogeneic immune responses and to improve graft survival. In this study, primary human hepatocytes (PHH) were isolated using a two-step-collagenase perfusion-technique and co-cultured with allogeneic lymphocytes in terms of a mixed lymphocyte hepatocyte culture. Expression of HLA class I on PHH was silenced using lentiviral vectors encoding for ß2-microglobulin-specific short hairpin RNA (shß2m) or non-specific shRNA (shNS) as control. The delivery of shß2m into PHH caused a decrease by up to 96% in ß2m transcript levels and a down-regulation of HLA class I cell surface expression on PHH by up to 57%. Proliferative T cell alloresponses against HLA-silenced PHH were significantly lower than those observed form fully HLA-expressing PHH. In addition, significantly lower secretion of pro-inflammatory cytokines was observed. Levels of albumin, urea and aspartate-aminotransferase did not differ in supernatants of cultured PHH. In conclusion, silencing HLA class I expression on PHH might represent a promising approach for immunomodulation in the transplant setting without compromising metabolic function of silenced hepatocytes.


Subject(s)
Gene Silencing , Hepatocytes/metabolism , Histocompatibility Antigens Class I/metabolism , Albumins/biosynthesis , Aspartate Aminotransferases/metabolism , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Humans , Killer Cells, Natural/metabolism , Ligands , Receptors, Cell Surface/metabolism , Urea/metabolism
7.
Hum Gene Ther ; 30(4): 485-496, 2019 04.
Article in English | MEDLINE | ID: mdl-30261752

ABSTRACT

Disparities at the major histocompatibility complex (MHC) antigens and associated minor antigens trigger harmful immune responses, leading to graft rejection after transplantation. We showed that MHC-silenced cells and tissues are efficiently protected against rejection. In complex vascularized organs, the endothelium is the major interface between donor and recipient. This study therefore aimed to reduce the immunogenicity of the lung by silencing MHC expression on the endothelium. In porcine lungs, short-hairpin RNAs targeting beta-2-microglobulin and class II-transactivator transcripts were delivered by lentiviral vectors during normothermic ex vivo perfusion to silence swine leukocyte antigen (SLA) I and II expression permanently. The results demonstrated the feasibility of genetically engineering all lung regions, achieving a targeted silencing effect for SLA I and II of 67% and 52%, respectively, without affecting cell viability or tissue integrity. This decrease in immunogenicity carries the potential to generate immunologically invisible organs to counteract the burden of rejection and immunosuppression.


Subject(s)
Endothelium, Vascular/metabolism , Gene Silencing , Genetic Engineering , Histocompatibility Antigens/genetics , Lung/metabolism , Animals , Endothelial Cells/metabolism , Gene Expression , Gene Transfer Techniques , Genes, Reporter , Genetic Vectors/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Lentivirus/genetics , Lung/pathology , Perfusion , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine , Temperature , Transduction, Genetic
8.
J Cell Mol Med ; 22(9): 4545-4549, 2018 09.
Article in English | MEDLINE | ID: mdl-29893509

ABSTRACT

Recently, platelet-derived growth factors present in lysates became an extreme interest in the field of regenerative medicine such as in wound healing and as substitutes to foetal bovine serum in xeno-free cell culture systems. However, the generation of such platelet lysates completely depends on the availability of platelet donors. In this study, the possibility to use in vitro-generated megakaryocytes derived from induced pluripotent stem cells (iPSCs) as a cell source for typical platelet growth factors was investigated. Therefore, the presence and levels of those factors were characterized in in vitro-produced megakaryocytes. In comparison with platelets, in vitro-generated megakaryocytes showed a multifold increased content in transcript and protein levels of typical platelet growth factors including platelet-derived growth factors (PDGFs), transforming growth factor (TGF)-1ß, vascular endothelial cell factor (VEGF)-A, epidermal growth factor (EGF), insulin-like growth factor (IGF)-1 and tissue factor (TF). Hence, iPSC-derived megakaryocytes may serve as an efficient cell source for a donor-independent generation of growth factor-rich lysates with a broad application potential in innovative cell culture systems and regenerative therapies.


Subject(s)
Gene Expression , Induced Pluripotent Stem Cells/cytology , Megakaryocytes/cytology , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Differentiation , Cell Extracts/chemistry , Cell Proliferation , Epidermal Growth Factor/biosynthesis , Epidermal Growth Factor/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Megakaryocytes/metabolism , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Primary Cell Culture , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Regenerative Medicine/methods , Thromboplastin/biosynthesis , Thromboplastin/genetics , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
9.
Biochim Biophys Acta ; 1853(10 Pt A): 2338-48, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26112988

ABSTRACT

The acidic nuclear phosphoproteins (ANP32A-H) are an evolutionarily conserved family of proteins with diverse and sometimes opposing cellular functions. Here we show that the oncogenic family members ANP32C and ANP32D are associated in complexes containing the molecular chaperone Hsp90. The oncogenic ANP32C protein appears to be highly unstable with a rapid degradation (t1/2>30 min) occurring upon treatment of cells with cycloheximide. ANP32C was also found to be associated with oncogenic Hsp90 complexes by virtue of its ability to interact and be immunoprecipitated by the Hsp90 inhibitor PU-H71. Further studies treating cells with the Hsp90 inhibitors PU-H71 and 17-AAG showed atypical increased protein stability and prevention of ANP32C degradation compared to the Hsp90 client AKT. Cells overexpressing ANP32C or its mutant ANP32CY140H showed enhanced sensitivity to treatment with PU-H71 as demonstrated by CCK-8 and colony formation assays. Our results highlight that certain malignancies with ANP32C/D overexpression or mutation might be specifically targeted using Hsp90 inhibitors.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Proteolysis , Amino Acid Substitution , Benzodioxoles/pharmacology , Cell Line , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Humans , Mutation, Missense , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein Stability/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Purines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...