Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 141(17): 2085-2099, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-36800642

RESUMO

Sickle cell disease (SCD) is a monogenic disease caused by a nucleotide mutation in the ß-globin gene. Current gene therapy studies are mainly focused on lentiviral vector-mediated gene addition or CRISPR/Cas9-mediated fetal globin reactivation, leaving the root cause unfixed. We developed a vectorized prime editing system that can directly repair the SCD mutation in hematopoietic stem cells (HSCs) in vivo in a SCD mouse model (CD46/Townes mice). Our approach involved a single intravenous injection of a nonintegrating, prime editor-expressing viral vector into mobilized CD46/Townes mice and low-dose drug selection in vivo. This procedure resulted in the correction of ∼40% of ßS alleles in HSCs. On average, 43% of sickle hemoglobin was replaced by adult hemoglobin, thereby greatly mitigating the SCD phenotypes. Transplantation in secondary recipients demonstrated that long-term repopulating HSCs were edited. Highly efficient target site editing was achieved with minimal generation of insertions and deletions and no detectable off-target editing. Because of its simplicity and portability, our in vivo prime editing approach has the potential for application in resource-poor countries where SCD is prevalent.


Assuntos
Anemia Falciforme , Edição de Genes , Camundongos , Animais , Edição de Genes/métodos , Sistemas CRISPR-Cas , Anemia Falciforme/genética , Anemia Falciforme/terapia , Células-Tronco Hematopoéticas , Hemoglobina Falciforme/genética
2.
Mol Ther ; 31(4): 1059-1073, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-36760126

RESUMO

We aim to develop an in vivo hematopoietic stem cell (HSC) gene therapy approach for persistent control/protection of HIV-1 infection based on the stable expression of a secreted decoy protein for HIV receptors CD4 and CCR5 (eCD4-Ig) from blood cells. HSCs in mice and a rhesus macaque were mobilized from the bone marrow and transduced by an intravenous injection of HSC-tropic, integrating HDAd5/35++ vectors expressing rhesus eCD4-Ig. In vivo HSC transduction/selection resulted in stable serum eCD4-Ig levels of ∼100 µg/mL (mice) and >20 µg/mL (rhesus) with half maximal inhibitory concentrations (IC50s) of 1 µg/mL measured by an HIV neutralization assay. After simian-human-immunodeficiency virus D (SHIV.D) challenge of rhesus macaques injected with HDAd-eCD4-Ig or a control HDAd5/35++ vector, peak plasma viral load levels were ∼50-fold lower in the eCD4-Ig-expressing animal. Furthermore, the viral load was lower in tissues with the highest eCD4-Ig expression, specifically the spleen and lymph nodes. SHIV.D challenge triggered a selective expansion of transduced CD4+CCR5+ cells, thereby increasing serum eCD4-Ig levels. The latter, however, broke immune tolerance and triggered anti-eCD4-Ig antibody responses, which could have contributed to the inability to eliminate SHIV.D. Our data will guide us in the improvement of the in vivo approach. Clearly, our conclusions need to be validated in larger animal cohorts.


Assuntos
Infecções por HIV , HIV-1 , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Humanos , Animais , Camundongos , Macaca mulatta , Vírus da Imunodeficiência Símia/genética , Células-Tronco Hematopoéticas , Síndrome de Imunodeficiência Adquirida dos Símios/terapia
3.
Blood ; 138(17): 1540-1553, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34086867

RESUMO

Thalassemia or sickle cell patients with hereditary persistence of fetal hemoglobin (HbF) have an ameliorated clinical phenotype and, in some cases, can achieve transfusion independence. Inactivation via genome editing of γ-globin developmental suppressors, such as BCL11A or LRF/ZBTB7A, or of their binding sites, have been shown to significantly increase expression of endogenous HbF. To broaden the therapeutic window beyond a single-editing approach, we have explored combinations of cis- and trans-editing targets to enhance HbF reactivation. Multiplex mutagenesis in adult CD34+ cells was well tolerated and did not lead to any detectable defect in the cells' proliferation and differentiation, either in vitro or in vivo. The combination of 1 trans and 1 cis mutation resulted in high editing retention in vivo, coupled with almost pancellular HbF expression in NBSGW mice. The greater in vivo performance of this combination was also recapitulated using a novel helper-dependent adenoviral-CRISPR vector (HD-Ad-dualCRISPR) in CD34+ cells from ß-thalassemia patients transplanted to NBSGW mice. A pronounced increase in HbF expression was observed in human red blood cells in mice with established predominant ß0/ß0-thalassemic hemopoiesis after in vivo injection of the HD-Ad-dualCRISPR vector. Collectively, our data suggest that the combination of cis and trans fetal globin reactivation mutations has the potential to significantly increase HbF both totally and on a per cell basis over single editing and could thus provide significant clinical benefit to patients with severe ß-globin phenotype.


Assuntos
Antígenos CD34/genética , Hemoglobina Fetal/genética , Mutagênese , Talassemia beta/genética , Adulto , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Edição de Genes , Terapia Genética , Humanos , Camundongos , Talassemia beta/terapia , gama-Globinas/genética
4.
Mol Ther ; 29(2): 822-837, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32949495

RESUMO

We have recently reported that, after in vivo hematopoietic stem cell/progenitor (HSPC) transduction with HDAd5/35++ vectors, SB100x transposase-mediated γ-globin gene addition achieved 10%-15% γ-globin of adult mouse globin, resulting in significant but incomplete phenotypic correction in a thalassemia intermedia mouse model. Furthermore, genome editing of a γ-globin repressor binding site within the γ-globin promoter by CRISPR-Cas9 results in efficient reactivation of endogenous γ-globin. Here, we aimed to combine these two mechanisms to obtain curative levels of γ-globin after in vivo HSPC transduction. We generated a HDAd5/35++ adenovirus vector (HDAd-combo) containing both modules and tested it in vitro and after in vivo HSPC transduction in healthy CD46/ß-YAC mice and in a sickle cell disease mouse model (CD46/Townes). Compared to HDAd vectors containing either the γ-globin addition or the CRISPR-Cas9 reactivation units alone, in vivo HSC transduction of CD46/Townes mice with the HDAd-combo resulted in significantly higher γ-globin in red blood cells, reaching 30% of that of adult human α and ßS chains and a complete phenotypic correction of sickle cell disease.


Assuntos
Adenoviridae/genética , Anemia Falciforme/genética , Anemia Falciforme/terapia , Terapia Genética , Vetores Genéticos/genética , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Animais , Modelos Animais de Doenças , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Camundongos , Transgenes
5.
Zentralbl Chir ; 147(1): 65-73, 2022 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-34225379

RESUMO

BACKGROUND: Funnel chest is a congenital deformity of the thorax in which the sternum and the adjacent ribs form a funnel towards the spine. As the pathogenesis has not yet been clearly clarified, there are different therapeutic approaches. These range from conservative measures and minimally invasive surgery to open corrective surgery. The patients suffer from aesthetic impairment, as well as cardiopulmonary restrictions due to the narrowing of the mediastinal space. The indication for funnel chest correction surgery is therefore justified by functional and aesthetic reasons. PATIENTS: We report a 23-year-old male patient who presented with subjectively impairing and objectively moderate funnel chest. A chest CT scan was performed preoperatively. The sternovertebral distance was 8 cm, the transverse thoracic diameter 28,9 cm (Haller's index 3.6). The preoperative pulmonary function test showed a slight restriction, the otherwise healthy patient described shortness of breath with greater exertion. The indication for funnel chest correction surgery was made. The operation was performed using a combined surgical technique: sternotomy and cartilage wedge resection according to Brunner/Grob and implantation of a metal bar without lateral antirotation plate. The most time-consuming step of the operation is the intraoperative adjustment of the metal bar. Therefore, in advance of the operation, we used a 3D printer to prepare a 3D reconstruction of the bony thorax with the help of the thorax CT scan. The metal bar was then bent and fitted to the thorax print and implanted during surgery. This allowed us to shorten the operation time by at least 15 min. RESULTS: The postoperative follow-up examinations showed a clearly erect funnel and a satisfactory aesthetic result. The metal bar explantation took place, as planned, 7 months after implantation. The postoperative CT scan now showed a Haller's index of 3.25, the pulmonary function test showed improved results. Subjectively, the patient was always symptom-free. CONCLUSION: The preparation of medical implants with 3D patient models as templates helps to save operation time. The practicability has not yet been established, as the creation of a 3D model of the bony thorax is associated with a different approach. With the increasing digitisation of the medical world, however, it is conceivable that the creation of digital and real 3D models will become easier and cheaper in the future.


Assuntos
Tórax em Funil , Adulto , Tórax em Funil/diagnóstico por imagem , Tórax em Funil/cirurgia , Humanos , Imageamento Tridimensional , Masculino , Procedimentos Cirúrgicos Minimamente Invasivos , Esterno/cirurgia , Tórax , Resultado do Tratamento , Adulto Jovem
6.
Blood ; 131(26): 2915-2928, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29789357

RESUMO

Disorders involving ß-globin gene mutations, primarily ß-thalassemia and sickle cell disease, represent a major target for hematopoietic stem/progenitor cell (HSPC) gene therapy. This includes CRISPR/Cas9-mediated genome editing approaches in adult CD34+ cells aimed toward the reactivation of fetal γ-globin expression in red blood cells. Because models involving erythroid differentiation of CD34+ cells have limitations in assessing γ-globin reactivation, we focused on human ß-globin locus-transgenic (ß-YAC) mice. We used a helper-dependent human CD46-targeting adenovirus vector expressing CRISPR/Cas9 (HDAd-HBG-CRISPR) to disrupt a repressor binding region within the γ-globin promoter. We transduced HSPCs from ß-YAC/human CD46-transgenic mice ex vivo and subsequently transplanted them into irradiated recipients. Furthermore, we used an in vivo HSPC transduction approach that involves HSPC mobilization and the intravenous injection of HDAd-HBG-CRISPR into ß-YAC/CD46-transgenic mice. In both models, we demonstrated efficient target site disruption, resulting in a pronounced switch from human ß- to γ-globin expression in red blood cells of adult mice that was maintained after secondary transplantation of HSPCs. In long-term follow-up studies, we did not detect hematological abnormalities, indicating that HBG promoter editing does not negatively affect hematopoiesis. This is the first study that shows successful in vivo HSPC genome editing by CRISPR/Cas9.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Globinas beta/genética , gama-Globinas/genética , Animais , Eritrócitos/metabolismo , Feminino , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas
7.
Mol Ther ; 27(12): 2195-2212, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31494053

RESUMO

Our goal is the development of in vivo hematopoietic stem cell (HSC) transduction technology with targeted integration. To achieve this, we modified helper-dependent HDAd5/35++ vectors to express a CRISPR/Cas9 specific to the "safe harbor" adeno-associated virus integration site 1 (AAVS1) locus and to provide a donor template for targeted integration through homology-dependent repair. We tested the HDAd-CRISPR + HDAd-donor vector system in AAVS1 transgenic mice using a standard ex vivo HSC gene therapy approach as well as a new in vivo HSC transduction approach that involves HSC mobilization and intravenous HDAd5/35++ injections. In both settings, the majority of treated mice had transgenes (GFP or human γ-globin) integrated into the AAVS1 locus. On average, >60% of peripheral blood cells expressed the transgene after in vivo selection with low-dose O6BG/bis-chloroethylnitrosourea (BCNU). Ex vivo and in vivo HSC transduction and selection studies with HDAd-CRISPR + HDAd-globin-donor resulted in stable γ-globin expression at levels that were significantly higher (>20% γ-globin of adult mouse globin) than those achieved in previous studies with a SB100x-transposase-based HDAd5/35++ system that mediates random integration. The ability to achieve therapeutically relevant transgene expression levels after in vivo HSC transduction and selection and targeted integration make our HDAd5/35++-based vector system a new tool in HSC gene therapy.


Assuntos
Adenoviridae/genética , Dependovirus/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Transdução Genética , Transgenes/fisiologia , Integração Viral , Animais , Sistemas CRISPR-Cas , Feminino , Genes Reporter , Terapia Genética , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , gama-Globinas/antagonistas & inibidores , gama-Globinas/genética
10.
Unfallchirurg ; 122(6): 452-463, 2019 Jun.
Artigo em Alemão | MEDLINE | ID: mdl-31165213

RESUMO

Surgical providers must maintain currency and competency for low-volume high-risk procedures for optimal outcomes. There are currently a number of methods available to train for these rare but vital skills, ranging from simple to complex and inexpensive to expensive. Traditionally, these skills have been taught using human cadaveric and animal models, which are limited by availability (cadavers) and social acceptability (animals). As such, there is a need to utilize advances in educational and simulation technologies to refine and develop consensus-based, validated, tissue-realistic, anatomically correct and cost-effective training tools to teach these vital skills. Partially perfused human cadavers have recently been shown to be an important adjunct to established trauma training. Human patient simulators (HPS) and the associated technology is rapidly expanding, but currently lack consistent realism to be used for competence training, when compared to traditional models and are currently cost-prohibitive. It will be important for surgical trainers and trainees to remain engaged and facilitate the development of realistic cost-effective training tools.


Assuntos
Necessidades e Demandas de Serviços de Saúde , Simulação de Paciente , Procedimentos Cirúrgicos Operatórios/educação , Animais , Competência Clínica , Simulação por Computador , Emergências , Humanos , Procedimentos Cirúrgicos Operatórios/normas , Inquéritos e Questionários
11.
J Virol ; 91(6)2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28077642

RESUMO

Defensins are small antimicrobial peptides capable of neutralizing human adenovirus (HAdV) in vitro by binding capsid proteins and blocking endosomal escape of virus. In humans, the alpha defensin HD5 is produced by specialized epithelial cells of the gastrointestinal and genito-urinary tracts. Here, we demonstrate, using patient biopsy specimens, that HD5 is also expressed as an active, secreted peptide by epithelial ovarian and lung cancer cells in situ This finding prompted us to study the role of HD5 in infection and spread of replication-competent, oncolytic HAdV type 3 (HAdV3). HAdV3 produces large amounts of penton-dodecahedra (PtDd), virus-like particles, during replication. We have previously shown that PtDd are involved in opening epithelial junctions, thus facilitating lateral spread of de novo-produced virions. Here, we describe a second function of PtDd, namely, the blocking of HD5. A central tool to prove that viral PtDd neutralize HD5 and support spread of progeny virus was an HAdV3 mutant virus in which formation of PtDd was disabled (mut-Ad3GFP, where GFP is green fluorescent protein). We demonstrated that viral spread of mut-Ad3GFP was blocked by synthetic HD5 whereas that of the wild-type (wt) form (wt-Ad3GFP) was only minimally impacted. In human colon cancer Caco-2 cells, induction of cellular HD5 expression by fibroblast growth factor 9 (FGF9) significantly inhibited viral spread and progeny virus production of mut-Ad3GFP but not of wt-Ad3GFP. Finally, the ectopic expression of HD5 in tumor cells diminished the in vivo oncolytic activity of mut-Ad3GFP but not of wt-Ad3GFP. These data suggest a new mechanism of HAdV3 to overcome innate antiviral host responses. Our study has implications for oncolytic adenovirus therapy.IMPORTANCE Previously, it has been reported that human defensin HD5 inactivates specific human adenoviruses by binding to capsid proteins and blocking endosomal escape of virus. The central new findings described in our manuscript are the following: (i) the discovery of a new mechanism used by human adenovirus serotype 3 to overcome innate antiviral host responses that is based on the capacity of HAdV3 to produce subviral penton-dodecahedral particles that act as decoys for HD5, thus preventing the inactivation of virus progeny produced upon replication; (ii) the demonstration that ectopic HD5 expression in cancer cells decreases the oncolytic efficacy of a serotype 5-based adenovirus vector; and (iii) the demonstration that epithelial ovarian and lung cancers express HD5. The study improves our understanding of how adenoviruses establish infection in epithelial tissues and has implications for cancer therapy with oncolytic adenoviruses.


Assuntos
Adenovírus Humanos/imunologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Evasão da Resposta Imune , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , alfa-Defensinas/metabolismo , Biópsia , Células CACO-2 , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Ovarianas/patologia
12.
Blood ; 128(18): 2206-2217, 2016 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-27554082

RESUMO

Current protocols for hematopoietic stem/progenitor cell (HSPC) gene therapy, involving the transplantation of ex vivo genetically modified HSPCs are complex and not without risk for the patient. We developed a new approach for in vivo HSPC transduction that does not require myeloablation and transplantation. It involves subcutaneous injections of granulocyte-colony-stimulating factor/AMD3100 to mobilize HSPCs from the bone marrow (BM) into the peripheral blood stream and the IV injection of an integrating, helper-dependent adenovirus (HD-Ad5/35++) vector system. These vectors target CD46, a receptor that is uniformly expressed on HSPCs. We demonstrated in human CD46 transgenic mice and immunodeficient mice with engrafted human CD34+ cells that HSPCs transduced in the periphery home back to the BM where they stably express the transgene. In hCD46 transgenic mice, we showed that our in vivo HSPC transduction approach allows for the stable transduction of primitive HSPCs. Twenty weeks after in vivo transduction, green fluorescent protein (GFP) marking in BM HSPCs (Lin-Sca1+Kit- cells) in most of the mice was in the range of 5% to 10%. The percentage of GFP-expressing primitive HSPCs capable of forming multilineage progenitor colonies (colony-forming units [CFUs]) increased from 4% of all CFUs at week 4 to 16% at week 12, indicating transduction and expansion of long-term surviving HSPCs. Our approach was well tolerated, did not result in significant transduction of nonhematopoietic tissues, and was not associated with genotoxicty. The ability to stably genetically modify HSPCs without the need of myeloablative conditioning is relevant for a broader clinical application of gene therapy.


Assuntos
Terapia Genética/métodos , Mobilização de Células-Tronco Hematopoéticas/métodos , Proteína Cofatora de Membrana/biossíntese , Transdução Genética/métodos , Adenoviridae , Animais , Vetores Genéticos/administração & dosagem , Células-Tronco Hematopoéticas , Xenoenxertos , Humanos , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL
13.
J Virol ; 89(21): 10841-59, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26292319

RESUMO

UNLABELLED: We recently discovered that desmoglein 2 (DSG2) is a receptor for human adenovirus species B serotypes Ad3, Ad7, Ad11, and Ad14. Ad3 is considered to be a widely distributed human pathogen. Ad3 binding to DSG2 triggers the transient opening of epithelial junctions. Here, we further delineate the mechanism that leads to DSG2-mediated epithelial junction opening in cells exposed to Ad3 and recombinant Ad3 fiber proteins. We identified an Ad3 fiber knob-dependent pathway that involves the phosphorylation of mitogen-activated protein (MAP) kinases triggering the activation of the matrix-metalloproteinase ADAM17. ADAM17, in turn, cleaves the extracellular domain of DSG2 that links epithelial cells together. The shed DSG2 domain can be detected in cell culture supernatant and also in serum of mice with established human xenograft tumors. We then extended our studies to Ad14 and Ad14P1. Ad14 is an important research and clinical object because of the recent appearance of a new, more pathogenic strain (Ad14P1). In a human epithelial cancer xenograft model, Ad14P1 showed more efficient viral spread and oncolysis than Ad14. Here, we tested the hypothesis that a mutation in the Ad14P1 fiber knob could account for the differences between the two strains. While our X-ray crystallography studies suggested an altered three-dimensional (3D) structure of the Ad14P1 fiber knob in the F-G loop region, this did not significantly change the fiber knob affinity to DSG2 or the intracellular signaling and DSG2 shedding in epithelial cancer cells. IMPORTANCE: A number of widely distributed adenoviruses use the epithelial junction protein DSG2 as a receptor for infection and lateral spread. Interaction with DSG2 allows the virus not only to enter cells but also to open epithelial junctions which form a physical barrier to virus spread. Our study elucidates the mechanism beyond virus-triggered junction opening with a focus on adenovirus serotype 3. Ad3 binds to DSG2 with its fiber knob domain and triggers intracellular signaling that culminates in the cleavage of the extracellular domain of DSG2, thereby disrupting DSG2 homodimers between epithelial cells. We confirmed this pathway with a second DSG2-interacting serotype, Ad14, and its recently emerged strain Ad14P1. These new insights in basic adenovirus biology can be employed to develop novel drugs to treat adenovirus infection as well as be used as tools for gene delivery into epithelial tissues or epithelial tumors.


Assuntos
Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Desmogleína 2/metabolismo , Modelos Moleculares , Proteínas ADAM/metabolismo , Proteína ADAM17 , Adenovírus Humanos/química , Análise de Variância , Animais , Western Blotting , Cristalografia por Raios X , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Fosforilação , Sorogrupo , Especificidade da Espécie , Ressonância de Plasmônio de Superfície , Espectrometria de Massas em Tandem
14.
Proc Natl Acad Sci U S A ; 110(40): 15919-24, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24046373

RESUMO

Most current cancer therapies focus on killing malignant cells, but these cells are often genetically unstable and can become resistant to chemotherapy. Tumor-associated macrophages (TAMs) facilitate disease progression by promoting angiogenesis and tumor cell growth, as well as by suppressing the adaptive immune response. TAMs are therefore potential targets for adjuvant anticancer therapies. However, resident macrophages are critical to host defense, and preferential ablation of TAMs remains challenging. Macrophage activation is broadly categorized as classically activated, or M1, and alternatively activated, or M2, and TAMs in the tumor microenvironment have been shown to adopt the anti-inflammatory, M2-like phenotype. To date, there are no methods for specific molecular targeting of TAMs. In this work, we report the discovery of a unique peptide sequence, M2pep, identified using a subtractive phage biopanning strategy against whole cells. The peptide preferentially binds to murine M2 cells, including TAMs, with low affinity for other leukocytes. Confocal imaging demonstrates the accumulation of M2pep in TAMs in vivo after tail vein injection. Finally, tail vein injection of an M2pep fusion peptide with a proapoptotic peptide delays mortality and selectively reduces the M2-like TAM population. This work therefore describes a molecularly targeted construct for murine TAMs and provides proof of concept of this approach as an anticancer treatment. In addition, M2pep is a useful tool for murine M2 macrophage identification and for modulating M2 macrophages in other murine models of disease involving M2 cells.


Assuntos
Apoptose/imunologia , Sistemas de Liberação de Medicamentos/métodos , Imunidade Inata/imunologia , Macrófagos/metabolismo , Neoplasias/imunologia , Peptídeos/metabolismo , Animais , Citometria de Fluxo , Macrófagos/imunologia , Camundongos , Microscopia Confocal , Biblioteca de Peptídeos , Peptídeos/imunologia , Análise de Sobrevida
15.
PLoS Pathog ; 9(10): e1003718, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204268

RESUMO

Human adenovirus serotypes Ad3, Ad7, Ad11, and Ad14 use the epithelial junction protein desmoglein 2 (DSG2) as a receptor for infection. During Ad infection, the fiber and penton base capsid proteins are produced in vast excess and form hetero-oligomers, called pentons. It has been shown for Ad3 that pentons self-assemble into penton-dodecahedra (PtDd). Our previous studies with recombinant purified Ad3 PtDd (produced in insect cells) showed that PtDd bind to DSG2 and trigger intracellular signaling resulting in the transient opening of junctions between epithelial cells. So far, a definitive proof for a function of Ad3 PtDd in the viral life cycle is elusive. Based on the recently published 3D structure of recombinant Ad3 PtDd, we generated a penton base mutant Ad3 vector (mu-Ad3GFP). mu-Ad3GFP is identical to its wild-type counterpart (wt-Ad3GFP) in the efficiency of progeny virus production; however, it is disabled in the production of PtDd. For infection studies we used polarized epithelial cancer cells or cell spheroids. We showed that in wt-Ad3GFP infected cultures, PtDd were released from cells before viral cytolysis and triggered the restructuring of epithelial junctions. This in turn facilitated lateral viral spread of de novo produced virions. These events were nearly absent in mu-Ad3GFP infected cultures. Our in vitro findings were consolidated in mice carrying xenograft tumors derived from human epithelial cancer cells. Furthermore, we provide first evidence that PtDd are also formed by another DSG2-interacting Ad serotype, the newly emerged, highly pathogenic Ad14 strain (Ad14p1). The central finding of this study is that a subgroup of Ads has evolved to generate PtDd as a strategy to achieve penetration into and dissemination in epithelial tissues. Our findings are relevant for basic and applied virology, specifically for cancer virotherapy.


Assuntos
Infecções por Adenovirus Humanos/transmissão , Adenovírus Humanos/metabolismo , Células Epiteliais/virologia , Junções Intercelulares/virologia , Vírion/metabolismo , Infecções por Adenovirus Humanos/genética , Infecções por Adenovirus Humanos/metabolismo , Adenovírus Humanos/genética , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células HeLa , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/patologia , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos
16.
Immunology ; 2014 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-25251258

RESUMO

The protein CD46 protects cells from complement attack by regulating cleavage of C3b and C3d. CD46 also regulates the adaptive immune response by controlling T cell activation and differentiation. Co-engagement of the T cell receptor and CD46 notably drives T cell differentiation by switching production of IFNγ to secretion of anti-inflammatory IL-10. This regulatory pathway is altered in several chronic inflammatory diseases highlighting its key role for immune homeostasis. The manipulation of the CD46 pathway may therefore provide a powerful means to regulate immune responses. Herein, we investigated the effect of recombinant proteins derived from the fiber knob of the adenovirus serotype 35 (Ad35) that uses CD46 as its entry receptor, on human T cell activation. We compared the effects of Ad35K++, engineered to exhibit enhanced affinity to CD46, and of Ad35K-, mutated in the binding site for CD46. Ad35K++ profoundly affects T cell activation by decreasing the levels of CD46 at the surface of primary T cells, and impairing T cell co-activation, shown by decreased CD25 expression, reduced proliferation and lower secretion of IL-10 and IFNγ. In contrast, Ad35K- acts a potent coactivator of T cells, enhancing T cell proliferation and cytokine production. These data show that recombinant Ad35 proteins are potent modulators of human T cell activation, and support their further development as potential drugs targeting T cell responses. This article is protected by copyright. All rights reserved.

17.
J Virol ; 87(21): 11346-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23946456

RESUMO

Human adenovirus (Ad) serotypes Ad3, Ad7, Ad11, and Ad14, as well as a recently emerged strain of Ad14 (Ad14p1), use the epithelial junction protein desmoglein 2 (DSG2) as a receptor for infection. Unlike Ad interaction with CAR and CD46, structural details for Ad binding to DSG2 are still elusive. Using an approach based on Escherichia coli expression libraries of random Ad3 and Ad14p1 fiber knob mutants, we identified amino acid residues that, when mutated individually, ablated or reduced Ad knob binding to DSG2. These residues formed three clusters inside one groove at the extreme distal end of the fiber knob. The Ad3 fiber knob mutant library was also used to identify variants with increased affinity to DSG2. We found a number of mutations within or near the EF loop of the Ad3 knob that resulted in affinities to DSG2 that were several orders of magnitude higher than those to the wild-type Ad3 knob. Crystal structure analysis of one of the mutants showed that the introduced mutations make the EF loop more flexible, which might facilitate the interaction with DSG2. Our findings have practical relevance for cancer therapy. We have recently reported that an Ad3 fiber knob-containing recombinant protein (JO-1) is able to trigger opening of junctions between epithelial cancer cells which, in turn, greatly improved the intratumoral penetration and efficacy of therapeutic agents (I. Beyer, et al., Clin. Cancer Res. 18:3340-3351, 2012; I. Beyer, et al., Cancer Res. 71:7080-7090, 2011). Here, we show that affinity-enhanced versions of JO-1 are therapeutically more potent than the parental protein in a series of cancer models.


Assuntos
Adenovírus Humanos/fisiologia , Proteínas do Capsídeo/metabolismo , Desmogleína 2/metabolismo , Interações Hospedeiro-Patógeno , Mapeamento de Interação de Proteínas , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular , Cristalografia por Raios X , Análise Mutacional de DNA , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores Virais/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
Mol Ther ; 21(2): 291-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23089733

RESUMO

We have developed a technology that depletes the complement regulatory protein (CRP) CD46 from the cell surface, and thereby sensitizes tumor cells to complement-dependent cytotoxicity triggered by therapeutic monoclonal antibodies (mAbs). This technology is based on a small recombinant protein, Ad35K++, which induces the internalization and subsequent degradation of CD46. In preliminary studies, we had demonstrated the utility of the combination of Ad35K++ and several commercially available mAbs such as rituximab, alemtuzumab, and trastuzumab in enhancing cell killing in vitro as well as in vivo in murine xenograft and syngeneic tumor models. We have completed scaled manufacturing of Ad35K++ protein in Escherichia coli for studies in nonhuman primates (NHPs). In macaques, we first defined a dose of the CD20-targeting mAb rituximab that did not deplete CD20-positive peripheral blood cells. Using this dose of rituximab, we then demonstrated that pretreatment with Ad35K++ reconstituted near complete elimination of B cells. Further studies demonstrated that the treatment was well tolerated and safe. These findings in a relevant large animal model provide the rationale for moving this therapy forward into clinical trials in patients with CD20-positive B-cell malignancies.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Linfócitos B/imunologia , Depleção Linfocítica , Proteína Cofatora de Membrana/genética , Alemtuzumab , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Murinos/imunologia , Antígenos CD20/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Macaca , Proteína Cofatora de Membrana/imunologia , Camundongos , Camundongos Transgênicos , Rituximab , Trastuzumab
19.
Cancer Biol Ther ; 25(1): 2314322, 2024 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-38361357

RESUMO

Multiple myeloma (MM) is an incurable malignancy of the B-cell lineage. Remarkable progress has been made in the treatment of MM with anti-CD38 monoclonal antibodies such as daratumumab and isatuximab, which can kill MM cells by inducing complement-dependent cytotoxicity (CDC). We showed that the CDC efficacy of daratumumab and isatuximab is limited by membrane complement inhibitors, including CD46 and CD59, which are upregulated in MM cells. We recently developed a small recombinant protein, Ad35K++, which is capable of transiently removing CD46 from the cell surface. We also produced a peptide inhibitor of CD59 (rILYd4). In this study, we tested Ad35K++ and rILYd4 in combination with daratumumab and isatuximab in MM cells as well as in cells from two other B-cell malignancies. We showed that Ad35K++ and rILYd4 increased CDC triggered by daratumumab and isatuximab. The combination of both inhibitors had an additive effect in vitro in primary MM cells as well as in vivo in a mouse xenograft model of MM. Daratumumab and isatuximab treatment of MM lines (without Ad35K++ or rILYd4) resulted in the upregulation of CD46/CD59 and/or survival of CD46high/CD59high MM cells that escaped the second round of daratumumab and isatuximab treatment. The escape in the second treatment cycle was prevented by the pretreatment of cells with Ad35K++. Overall, our data demonstrate that Ad35K++ and rILYd4 are efficient co-therapeutics of daratumumab and isatuximab, specifically in multi-cycle treatment regimens, and could be used to improve treatment of multiple myeloma.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Humanos , Camundongos , Animais , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/uso terapêutico , ADP-Ribosil Ciclase 1/metabolismo , Antígenos CD59/uso terapêutico , Proteína Cofatora de Membrana/metabolismo
20.
Blood Adv ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38843380

RESUMO

We have reported direct repair of the sickle cell mutation in vivo in a disease model using vectorized prime editors after hematopoietic stem cell (HSC) mobilization with G-CSF/AMD3100. The use of G-CSF for HSC mobilization would be a hurdle for the clinical translation of the approach. Here, we tested a G-CSF-free mobilization regimen using WU-106, a PEG-conjugated inhibitor of integrin VLA-4 (4ß1), plus AMD3100 for in vivo HSC prime editing in sickle cell disease (SCD) mice (CD46/Townes). Mobilization with WU-106+AMD3100 in CD46/Townes mice was rapid and efficient. In contrast to the G-CSF/AMD3100 approach, mobilization of activated granulocytes and elevation of the key pro-inflammatory cytokine IL-6 in serum were minimal. The combination of WU-106+AMD3100 mobilization and intravenous injection of an HDAd-PE5 vector together with in vivo selection resulted in a SCD mutation editing (T>A correction) rate of ~23% in bone marrow and peripheral blood cells of CD46/Townes mice. The treated mice demonstrated phenotypic correction, reflected by normalized blood parameters and spleen size. Editing rates were significantly increased (29%) in secondary recipients indicating preferential mobilization/transduction of long-term repopulating HSCs. Using this approach, we found <1% of undesired indels and no detectable off-target editing at top-scored potential sites. Our study shows that in vivo transduction to treat SCD (including HSC mobilization and HDAd injection) can now be done within 2 hours involving only simple intravenous injections with a good safety profile. The same-day mobilization regimen makes in vivo HSC gene therapy more attractive for the resource-poor settings where SCD does the most damage.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA