Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Haematologica ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38988258

ABSTRACT

Umbilical cord blood (UCB) T cells exhibit distinct naive ontogenetic profiles and may be an attractive source of starting cells for the production of chimeric antigen receptor (CAR) T cells. Pre-selection of UCB-T cells on the basis of CD62L expression was investigated as part of a machine-based manufacturing process, incorporating lentiviral transduction, CRISPR-Cas9 editing, T-cell expansion and depletion of residual TCReeeT cells. This provided stringent mitigation against the risk of graft versus host disease (GVHD), and was combined with simultaneous knockout of CD52 to enable persistence of edited T cells in combination with preparative lymphodepletion using Alemtuzumab. Under compliant manufacturing conditions, two cell banks were generated with high levels of CAR19 expression and minimal carriage of TCReeeT cells. Sufficient cells were cryopreserved in dose-banded aliquots at the end of each campaign to treat dozens of potential recipients. Molecular characterisation captured vector integration sites and CRISPR editing signatures and functional studies, including in vivo potency studies in humanised mice, confirmed antileukaemic activity comparable to peripheral blood-derived universal CAR19 T cells. Machine manufactured UCB derived T cells banks offer an alternative to autologous cell therapies and could help widen access to CAR T cells.

2.
N Engl J Med ; 389(10): 899-910, 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37314354

ABSTRACT

BACKGROUND: Cytidine deamination that is guided by clustered regularly interspaced short palindromic repeats (CRISPR) can mediate a highly precise conversion of one nucleotide into another - specifically, cytosine to thymine - without generating breaks in DNA. Thus, genes can be base-edited and rendered inactive without inducing translocations and other chromosomal aberrations. The use of this technique in patients with relapsed childhood T-cell leukemia is being investigated. METHODS: We used base editing to generate universal, off-the-shelf chimeric antigen receptor (CAR) T cells. Healthy volunteer donor T cells were transduced with the use of a lentivirus to express a CAR with specificity for CD7 (CAR7), a protein that is expressed in T-cell acute lymphoblastic leukemia (ALL). We then used base editing to inactivate three genes encoding CD52 and CD7 receptors and the ß chain of the αß T-cell receptor to evade lymphodepleting serotherapy, CAR7 T-cell fratricide, and graft-versus-host disease, respectively. We investigated the safety of these edited cells in three children with relapsed leukemia. RESULTS: The first patient, a 13-year-old girl who had relapsed T-cell ALL after allogeneic stem-cell transplantation, had molecular remission within 28 days after infusion of a single dose of base-edited CAR7 (BE-CAR7). She then received a reduced-intensity (nonmyeloablative) allogeneic stem-cell transplant from her original donor, with successful immunologic reconstitution and ongoing leukemic remission. BE-CAR7 cells from the same bank showed potent activity in two other patients, and although fatal fungal complications developed in one patient, the other patient underwent allogeneic stem-cell transplantation while in remission. Serious adverse events included cytokine release syndrome, multilineage cytopenia, and opportunistic infections. CONCLUSIONS: The interim results of this phase 1 study support further investigation of base-edited T cells for patients with relapsed leukemia and indicate the anticipated risks of immunotherapy-related complications. (Funded by the Medical Research Council and others; ISRCTN number, ISRCTN15323014.).


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adolescent , Child , Female , Humans , Antigens, CD19 , Antigens, CD7 , CD52 Antigen , Hematopoietic Stem Cell Transplantation/adverse effects , Immunotherapy, Adoptive/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/genetics , Recurrence , Stem Cell Transplantation , T-Lymphocytes
3.
Hematol Oncol Stem Cell Ther ; 15(3): 137-152, 2022 Nov 07.
Article in English | MEDLINE | ID: mdl-36395497

ABSTRACT

Increasing success of adaptive cell therapy (ACT), such as genetically engineered T cells to express chimeric antigen receptors (CARs) proven to be highly significant technological advancements and impressive clinical outcomes in selected haematological malignancies, with promising efficacy. The evolution of CAR designs beyond the conventional structures is necessary to address some of the limitations of conventional CAR therapy and to expand the use of CAR T cells to a wider range of malignancies. There are various obstacles with a wide range of engineering strategies in order to improve the safety, efficacy and applicability of this therapeutic modality. Here we describe details of modular CAR structure with all the necessary domains and what is known about proximal CAR signalling in T cells. Furthermore, the global need for adoptive cell therapy is expanding very rapidly, and there is an urgent increasing demand for fully automated manufacturing methods that can produce large scale clinical grade high quality CAR engineered immune cells. Despite the advances in automation for the production of clinical grade CAR engineered cells, the manufacturing process is costly, consistent and involves multiple steps, including selection, activation, transduction, and Ex-Vivo expansion. Among these complex manufacturing phases, the choice of culture system to generate a high number of functional cells needs to be evaluated and optimized. Here we list the most advance fully automated to semi-automated bioreactor platforms can be used for the production of clinical grade CAR engineered cells for clinical trials but are far from being standardized. New processing options are available and a systematic effort seeking automation, standardization and the increase of production scale, would certainly help to bring the costs down and ultimately democratise this personalized therapy. In this review, we describe in detail different CAR engineered T cell platforms available and can be used in future for clinical-grade CAR engineered ATMP production.


Subject(s)
Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , T-Lymphocytes , Cell- and Tissue-Based Therapy , Bioreactors
4.
Sci Transl Med ; 14(668): eabq3010, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36288281

ABSTRACT

Genome editing of allogeneic T cells can provide "off-the-shelf" alternatives to autologous chimeric antigen receptor (CAR) T cell therapies. Disruption of T cell receptor α chain (TRAC) to prevent graft-versus-host disease (GVHD) and removal of CD52 (cluster of differentiation 52) for a survival advantage in the presence of alemtuzumab have previously been investigated using transcription activator-like effector nuclease (TALEN)-mediated knockout. Here, we deployed next-generation CRISPR-Cas9 editing and linked CAR expression to multiplexed DNA editing of TRAC and CD52 through incorporation of self-duplicating CRISPR guide RNA expression cassettes within the 3' long terminal repeat of a CAR19 lentiviral vector. Three cell banks of TT52CAR19 T cells were generated and cryopreserved. A phase 1, open-label, non-randomized clinical trial was conducted and treated six children with relapsed/refractory CD19-positive B cell acute lymphoblastic leukemia (B-ALL) (NCT04557436). Lymphodepletion included fludarabine, cyclophosphamide, and alemtuzumab and was followed by a single infusion of 0.8 × 106 to 2.0 × 106 CAR19 T cells per kilogram with no immediate toxicities. Four of six patients infused with TT52CAR19 T cells exhibited cell expansion, achieved flow cytometric remission, and then proceeded to receive allogeneic stem cell transplantation. Two patients required biological intervention for grade II cytokine release syndrome, one patient developed transient grade IV neurotoxicity, and one patient developed skin GVHD, which resolved after transplant conditioning. Other complications were within expectations, and primary safety objectives were met. This study provides a demonstration of the feasibility, safety, and therapeutic potential of CRISPR-engineered immunotherapy.


Subject(s)
Graft vs Host Disease , Leukemia, B-Cell , Leukemia, Lymphocytic, Chronic, B-Cell , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Child , Humans , Alemtuzumab , Antigens, CD19/metabolism , Cyclophosphamide , Graft vs Host Disease/metabolism , Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Chimeric Antigen/metabolism , RNA, Guide, Kinetoplastida/metabolism , T-Lymphocytes , Transcription Activator-Like Effector Nucleases/genetics
5.
Adv Funct Mater ; 31(37): 2104843, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-35712226

ABSTRACT

The authors aim to develop siRNA therapeutics for cancer that can be administered systemically to target tumors and retard their growth. The efficacy of systemic delivery of siRNA to tumors with nanoparticles based on lipids or polymers is often compromised by their rapid clearance from the circulation by the liver. Here, multifunctional cationic and anionic siRNA nanoparticle formulations are described, termed receptor-targeted nanocomplexes (RTNs), that comprise peptides for siRNA packaging into nanoparticles and receptor-mediated cell uptake, together with lipids that confer nanoparticles with stealth properties to enhance stability in the circulation, and fusogenic properties to enhance endosomal release within the cell. Intravenous administration of RTNs in mice leads to predominant accumulation in xenograft tumors, with very little detected in the liver, lung, or spleen. Although non-targeted RTNs also enter the tumor, cell uptake appears to be RGD peptide-dependent indicating integrin-mediated uptake. RTNs with siRNA against MYCN (a member of the Myc family of transcription factors) in mice with MYCN-amplified neuroblastoma tumors show significant retardation of xenograft tumor growth and enhanced survival. This study shows that RTN formulations can achieve specific tumor-targeting, with minimal clearance by the liver and so enable delivery of tumor-targeted siRNA therapeutics.

6.
Hum Gene Ther ; 30(9): 1067-1078, 2019 09.
Article in English | MEDLINE | ID: mdl-31288584

ABSTRACT

Netherton syndrome (NS) is a rare autosomal recessive skin disorder caused by mutations in SPINK5. It is a debilitating condition with notable mortality in the early years of life. There is no curative treatment. We undertook a nonrandomized, open-label, feasibility, and safety study using autologous keratinocytes transduced with a lentiviral vector encoding SPINK5 under the control of the human involucrin promoter. Six NS subjects were recruited, and gene-modified epithelial sheets were successfully generated in three of five subjects. The sheets exhibited expression of correctly sized lympho-epithelial Kazal-type-related inhibitor (LEKTI) protein after modification. One subject was grafted with a 20 cm2 gene-modified graft on the left anterior thigh without any adverse complications and was monitored by serial sampling for 12 months. Recovery within the graft area was compared against an area outside by morphology, proviral copy number and expression of the SPINK5 encoded protein, LEKTI, and its downstream target kallikrein 5, which exhibited transient functional correction. The study confirmed the feasibility of generating lentiviral gene-modified epidermal sheets for inherited skin diseases such as NS, but sustained LEKTI expression is likely to require the identification, targeting, and engraftment of long-lived keratinocyte stem cell populations for durable therapeutic effects. Important learning points for the application of gene-modified epidermal sheets are discussed.


Subject(s)
Epidermal Cells/metabolism , Epidermis/metabolism , Epidermis/transplantation , Netherton Syndrome/genetics , Netherton Syndrome/therapy , Transduction, Genetic , Transgenes , Adolescent , Adult , Autografts , Biomarkers , Cell Culture Techniques , Female , Fluorescent Antibody Technique , Gene Expression , Genetic Engineering , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Immunohistochemistry , Keratinocytes/metabolism , Lentivirus/genetics , Male , Mutation , Netherton Syndrome/metabolism , Netherton Syndrome/pathology , Serine Peptidase Inhibitor Kazal-Type 5/genetics , Serine Peptidase Inhibitor Kazal-Type 5/metabolism , Treatment Outcome , Young Adult
7.
JCI Insight ; 4(11)2019 06 06.
Article in English | MEDLINE | ID: mdl-31167965

ABSTRACT

BACKGROUNDRecessive dystrophic epidermolysis bullosa (RDEB) is a severe form of skin fragility disorder due to mutations in COL7A1 encoding basement membrane type VII collagen (C7), the main constituent of anchoring fibrils (AFs) in skin. We developed a self-inactivating lentiviral platform encoding a codon-optimized COL7A1 cDNA under the control of a human phosphoglycerate kinase promoter for phase I evaluation.METHODSIn this single-center, open-label phase I trial, 4 adults with RDEB each received 3 intradermal injections (~1 × 106 cells/cm2 of intact skin) of COL7A1-modified autologous fibroblasts and were followed up for 12 months. The primary outcome was safety, including autoimmune reactions against recombinant C7. Secondary outcomes included C7 expression, AF morphology, and presence of transgene in the injected skin.RESULTSGene-modified fibroblasts were well tolerated, without serious adverse reactions or autoimmune reactions against recombinant C7. Regarding efficacy, there was a significant (P < 0.05) 1.26-fold to 26.10-fold increase in C7 mean fluorescence intensity in the injected skin compared with noninjected skin in 3 of 4 subjects, with a sustained increase up to 12 months in 2 of 4 subjects. The presence of transgene (codon-optimized COL7A1 cDNA) was demonstrated in the injected skin at month 12 in 1 subject, but no new mature AFs were detected.CONCLUSIONTo our knowledge, this is the first human study demonstrating safety and potential efficacy of lentiviral fibroblast gene therapy with the presence of COL7A1 transgene and subsequent C7 restoration in vivo in treated skin at 1 year after gene therapy. These data provide a rationale for phase II studies for further clinical evaluation.TRIAL REGISTRATIONClincalTrials.gov NCT02493816.FUNDINGCure EB, Dystrophic Epidermolysis Bullosa Research Association (UK), UK NIHR Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, and Fondation René Touraine Short-Exchange Award.


Subject(s)
Epidermolysis Bullosa Dystrophica/therapy , Fibroblasts , Genetic Therapy , Lentivirus/genetics , Adult , Collagen Type VII/genetics , Female , Fibroblasts/metabolism , Fibroblasts/transplantation , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Male , Middle Aged , Treatment Outcome
9.
J Invest Dermatol ; 136(1): 284-92, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26763448

ABSTRACT

Cells therapies, engineered to secrete replacement proteins, are being developed to ameliorate otherwise debilitating diseases. Recessive dystrophic epidermolysis bullosa (RDEB) is caused by defects of type VII collagen, a protein essential for anchoring fibril formation at the dermal-epidermal junction. Whereas allogeneic fibroblasts injected directly into the dermis can mediate transient disease modulation, autologous gene-modified fibroblasts should evade immunological rejection and support sustained delivery of type VII collagen at the dermal-epidermal junction. We demonstrate the feasibility of such an approach using a therapeutic grade, self-inactivating-lentiviral vector, encoding codon-optimized COL7A1, to transduce RDEB fibroblasts under conditions suitable for clinical application. Expression and secretion of type VII collagen was confirmed with transduced cells exhibiting supranormal levels of protein expression, and ex vivo migration of fibroblasts was restored in functional assays. Gene-modified RDEB fibroblasts also deposited type VII collagen at the dermal-epidermal junction of human RDEB skin xenografts placed on NOD-scid IL2Rgamma(null) recipients, with reconstruction of human epidermal structure and regeneration of anchoring fibrils at the dermal-epidermal junction. Fibroblast-mediated restoration of protein and structural defects in this RDEB model strongly supports proposed therapeutic applications in man.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/therapy , Fibroblasts/transplantation , Animals , Codon , Disease Models, Animal , Gene Expression Regulation , Genetic Vectors , Heterografts , Humans , Lentivirus/genetics , Male , Mice , Mice, SCID , Random Allocation , Skin Transplantation/methods , Tissue Engineering , Wound Healing/physiology
10.
Blood ; 126(13): 1527-35, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26224645

ABSTRACT

Megakaryoblastic leukemia 1 (MKL1), also known as MAL or myocardin-related transcription factor A (MRTF-A), is a coactivator of serum response factor, which regulates transcription of actin and actin cytoskeleton-related genes. MKL1 is known to be important for megakaryocyte differentiation and function in mice, but its role in immune cells is unexplored. Here we report a patient with a homozygous nonsense mutation in the MKL1 gene resulting in immunodeficiency characterized predominantly by susceptibility to severe bacterial infection. We show that loss of MKL1 protein expression causes a dramatic loss of filamentous actin (F-actin) content in lymphoid and myeloid lineage immune cells and widespread cytoskeletal dysfunction. MKL1-deficient neutrophils displayed reduced phagocytosis and almost complete abrogation of migration in vitro. Similarly, primary dendritic cells were unable to spread normally or to form podosomes. Silencing of MKL1 in myeloid cell lines revealed that F-actin assembly was abrogated through reduction of globular actin (G-actin) levels and disturbed expression of multiple actin-regulating genes. Impaired migration of these cells was associated with failure of uropod retraction likely due to altered contractility and adhesion, evidenced by reduced expression of the myosin light chain 9 (MYL9) component of myosin II complex and overexpression of CD11b integrin. Together, our results show that MKL1 is a nonredundant regulator of cytoskeleton-associated functions in immune cells and fibroblasts and that its depletion underlies a novel human primary immunodeficiency.


Subject(s)
Codon, Nonsense , Immunologic Deficiency Syndromes/genetics , Pseudomonas Infections/genetics , Trans-Activators/genetics , Actins/metabolism , Actins/ultrastructure , Cell Line , Cell Movement , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Homozygote , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Pseudomonas/isolation & purification , Pseudomonas Infections/complications , Pseudomonas Infections/diagnosis , Pseudomonas Infections/metabolism
11.
PLoS One ; 10(4): e0123016, 2015.
Article in English | MEDLINE | ID: mdl-25849484

ABSTRACT

Notch signalling is critical for haemopoietic stem cell (HSC) self-renewal and survival. The role of Notch signalling has been reported recently in chronic myeloid leukaemia (CML) - a stem cell disease characterized by BCR-ABL tyrosine kinase activation. Therefore, we studied the relationship between BCR-ABL and Notch signalling and assessed the expression patterns of Notch and its downstream target Hes1 in CD34+ stem and progenitor cells from chronic-phase CML patients and bone marrow (BM) from normal subjects (NBM). We found significant upregulation (p<0.05) of Notch1, Notch2 and Hes1 on the most primitive CD34+Thy+ subset of CML CD34+ cells suggesting that active Notch signalling in CML primitive progenitors. In addition, Notch1 was also expressed in distinct lymphoid and myeloid progenitors within the CD34+ population of primary CML cells. To further delineate the possible role and interactions of Notch with BCR-ABL in CD34+ primary cells from chronic-phase CML, we used P-crkl detection as a surrogate assay of BCR-ABL tyrosine kinase activity. Our data revealed that Imatinib (IM) induced BCR-ABL inhibition results in significant (p<0.05) upregulation of Notch activity, assessed by Hes1 expression. Similarly, inhibition of Notch leads to hyperactivation of BCR-ABL. This antagonistic relationship between Notch and BCR-ABL signalling was confirmed in K562 and ALL-SIL cell lines. In K562, we further validated this antagonistic relationship by inhibiting histone deacetylase (HDAC) - an effector pathway of Hes1, using valproic acid (VPA) - a HDAC inhibitor. Finally, we also confirmed the potential antagonism between Notch and BCR/ABL in In Vivo, using publically available GSE-database, by analysing gene expression profile of paired samples from chronic-phase CML patients pre- and post-Imatinib therapy. Thus, we have demonstrated an antagonistic relationship between Notch and BCR-ABL in CML. A combined inhibition of Notch and BCR-ABL may therefore provide superior clinical response over tyrosine-kinase inhibitor monotherapy by targeting both quiescent leukaemic stem cells and differentiated leukaemic cells and hence must be explored.


Subject(s)
Antigens, CD34/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Fusion Proteins, bcr-abl/metabolism , Homeodomain Proteins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Fusion Proteins, bcr-abl/genetics , Homeodomain Proteins/genetics , Humans , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/therapeutic use , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor HES-1 , Tumor Cells, Cultured
12.
Arch Dermatol Res ; 307(2): 115-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25322916

ABSTRACT

Among raised dermal scar types, keloid (KS) and hypertrophic scars (HS) are considered to present clinical similarities, but there are no known specific biomarkers that allow both scar types to be easily distinguished. Development and progression of raised dermal scars comprises the activation of several molecular pathways and cell defence mechanisms leading to elevated extracellular matrix component synthesis, delayed apoptosis, altered migration and differentiation. Therefore, the aim here was to identify biomarkers that may differentiate between KS and HS compared to normal skin (NS). To achieve this aim, NS (n = 14), KS (n = 14) and HS (n = 14) biopsies were evaluated using histology by H&E staining. Tissue biopsies and primary fibroblasts (passages 0-4) were employed to assess the gene expression levels of 21 biomarkers selected from our previous microarray studies using qRT-PCR. Finally, protein expression was evaluated using In-Cell Western Blotting in primary fibroblasts (p 0-4). Our results demonstrated that out of the 21 biomarkers screened at mRNA and protein levels, α2ß1-integrin, Hsp27, PAI-2, MMP-19 and CGRP showed significantly higher expression (p < 0.05) in KS compared to NS and HS. Additionally, these five key biomarkers were found to be significantly higher (p < 0.05) at mRNA level in KS taken from the sternum, a region known to be subjected to high mechanical forces in the body during the performance of daily movements. In conclusion, our findings offer potential molecular targets in raised dermal scars differentiation. Future targeted research may allow provision of diagnostic and prognostic markers in keloid versus hypertrophic scars.


Subject(s)
Biomarkers/analysis , Cicatrix, Hypertrophic/diagnosis , Gene Expression Profiling , Keloid/diagnosis , Blotting, Western , Cicatrix, Hypertrophic/genetics , Humans , Keloid/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
13.
Wound Repair Regen ; 22(5): 557-68, 2014.
Article in English | MEDLINE | ID: mdl-25046655

ABSTRACT

Skin tension may influence keloid scar behavior, development, and spreading, e.g., butterfly-shaped keloid disease in the sternum. Here, we developed a three-dimensional (3D) in vitro model to mimic in vivo tension and evaluate keloid fibroblast (KF) behavior and extracellular matrix synthesis under tension. In vivo skin tension measured in volunteers (n = 4) using 3D image photogrammetry enabled prediction of actual force (35 mN). A novel cell force monitor applied tension in a fibroblast-populated 3D collagen lattice replicating the in vivo force. The effect of tension on keloid (n = 10) fibroblast (KF) and normal skin (n = 10) fibroblasts (NF) at set time points (6, 12, and 24 hours) was measured in Hsp27, PAI-2, and α2ß1 integrin, tension-related genes demonstrating significant (p < 0.05) time-dependent regulation of these genes in NF vs. KF with and without tension. KF showed higher (p < 0.05) proliferation post-tension. Knockdown of all three genes in 24 and 48 hours with and without tension showed significant down-regulation in NF vs. KF. Additionally, we show significant (p < 0.05) modification of the expression of extracellular matrix-related genes post-tension following down-regulation of Hsp27, PAI-2, or α2ß1 integrin. Finally, we demonstrate significant alteration in NF compared with KF morphology following knockdown. In conclusion, this study shows induction of tension-related genes expression following mechano-regulation in KFs, with potential relevance to its development and therapy.


Subject(s)
Fibroblasts/metabolism , HSP27 Heat-Shock Proteins/genetics , Integrin alpha2beta1/genetics , Keloid/genetics , Plasminogen Activator Inhibitor 2/genetics , RNA, Messenger/metabolism , Stress, Mechanical , Wound Healing/genetics , Adolescent , Adult , Aged , Case-Control Studies , Extracellular Matrix/metabolism , Female , Fibroblasts/pathology , Gene Expression Regulation , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Integrin alpha2beta1/metabolism , Keloid/metabolism , Keloid/pathology , Male , Middle Aged , Molecular Chaperones , Plasminogen Activator Inhibitor 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Wound Healing/physiology , Young Adult
14.
PLoS One ; 8(12): e75600, 2013.
Article in English | MEDLINE | ID: mdl-24348987

ABSTRACT

Keloid disease (KD) is an abnormal cutaneous fibroproliferative disorder of unknown aetiopathogenesis. Keloid fibroblasts (KF) are implicated as mediators of elevated extracellular matrix deposition. Aberrant secretory behaviour by KF relative to normal skin fibroblasts (NF) may influence the disease state. To date, no previous reports exist on the ability of site-specific KF to induce fibrotic-like phenotypic changes in NF or normal scar fibroblasts (NS) by paracrine mechanisms. Therefore, the aim of this study was to investigate the influence of conditioned media from site-specific KF on the cellular and molecular behaviour of both NF and NS enabled by paracrine mechanisms. Conditioned media was collected from cultured primary fibroblasts during a proliferative log phase of growth including: NF, NS, peri-lesional keloid fibroblasts (PKF) and intra-lesional keloid fibroblasts (IKF). Conditioned media was used to grow NF, NS, PKF and IKF cells over 240 hrs. Cellular behavior was monitored through real time cell analysis (RTCA), proliferation rates and migration in a scratch wound assay. Fibrosis-associated marker expression was determined at both protein and gene level. PKF conditioned media treatment of both NF and NS elicited enhanced cell proliferation, spreading and viability as measured in real time over 240 hrs versus control conditioned media. Following PKF and IKF media treatments up to 240 hrs, both NF and NS showed significantly elevated proliferation rates (p<0.03) and migration in a scratch wound assay (p<0.04). Concomitant up-regulation of collagen I, fibronectin, α-SMA, PAI-1, TGF-ß and CTGF (p<0.03) protein expression were also observed. Corresponding qRT-PCR analysis supported these findings (P<0.03). In all cases, conditioned media from growing marginal PKF elicited the strongest effects. In conclusion, primary NF and NS cells treated with PKF or IKF conditioned media exhibit enhanced expression of fibrosis-associated molecular markers and increased cellular activity as a result of keloid fibroblast-derived paracrine factors.


Subject(s)
Cicatrix/metabolism , Cicatrix/pathology , Fibroblasts/cytology , Fibroblasts/metabolism , Skin/cytology , Skin/pathology , Adult , Aged , Blotting, Western , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Female , Fibroblasts/drug effects , Humans , Keloid , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism
15.
Lab Invest ; 93(8): 946-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23835737

ABSTRACT

Keloid disease (KD) is a common fibroproliferative disorder of unknown etiopathogenesis. Its unique occurrence in human skin and lack of animal models pose challenges for KD research. The lack of a suitable model in KD and over-reliance on cell culture has hampered the progress in developing new treatments. Therefore, we evaluated the effect of two promising candidate antifibrotic therapies: (-)-epigallocatechin-3-gallate (EGCG) and plasminogen activator inhibitor-1 (PAI-1) silencing in a long-term human keloid organ culture (OC). Four millimeters of air-liquid interface (ALI) keloid explants on collagen gel matrix in serum-free medium (n=8 cases) were treated with different modalities (EGCG treatment; PAI-1 knockdown by short interfering RNA (siRNA) and application of dexamethasone (DEX) as control). Normal skin (n=6) was used as control (only for D0 keloid-untreated comparison). Besides routine histology and quantitative (immuno-) histomorphometry, the key phenotypic and growth parameters of KD were assessed. Results demonstrated that EGCG reduced keloid volume significantly (40% by week 4), increased apoptosis (≥40% from weeks 1 to 4), and decreased proliferation (≤17% in week 2). EGCG induced epidermal shrinkage, reduced collagen-I and -III at mRNA and protein levels, depleted 98% of keloid-associated mast cells, and reduced the percentage of both cellularity and blood vessel count by week 4. Knockdown of PAI-1 significantly reduced keloid volume by 28% in week 4, respectively, and reduced collagen-I and -III at both mRNA and protein levels. As expected, DEX increased keloid apoptosis, decreased keloid proliferation, and collagen synthesis, but induced connective tissue growth factor overexpression. In conclusion, using keloid OC model, we provide the first functional evidence for testing candidate antifibrotic compounds in KD. We show that EGCG and PAI-1 silencing effectively inhibits growth and induces shrinkage of human keloid tissue in situ. Therefore, the application of EGCG, PAI-1 silencing, and other emerging compounds tested using this model may provide effective treatment and potentially aid in the prevention of recurrence of KD following surgery.


Subject(s)
Catechin/analogs & derivatives , Gene Silencing , Keloid/drug therapy , Plasminogen Activator Inhibitor 1/genetics , Skin/drug effects , Adolescent , Adult , Apoptosis/drug effects , Apoptosis/genetics , Catechin/pharmacology , Cell Proliferation/drug effects , Collagen/metabolism , Dexamethasone/pharmacology , Gene Knockdown Techniques , Humans , Keloid/genetics , Keloid/pathology , Organ Culture Techniques , RNA, Small Interfering/genetics , Skin/metabolism , Skin/pathology , Young Adult
16.
Plast Reconstr Surg ; 131(2): 158e-173e, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23358011

ABSTRACT

BACKGROUND: Keloid disease is a fibroproliferative disorder, with an ill-defined treatment that is characterized by excessive extracellular matrix deposition. Mechanical tension promotes deposition of extracellular matrix and overexpression of tension-related proteins, which is associated with keloid disease. The aim of this study was to investigate the effect of tension-related proteins on extracellular matrix steady-state synthesis in primary keloid fibroblasts. METHODS: Keloid fibroblasts (n = 10) and normal skin (n = 4) fibroblast cultures were established from passages 0 to 3. A panel of 21 tension-related genes from microarray data were assessed at mRNA (quantitative reverse-transcriptase polymerase chain reaction) and protein (in-cell Western blotting) levels. Three genes were significantly altered in keloid tissue and fibroblasts, and their functional role was assessed using siRNA knockdown. RESULTS: Hsp27, α2ß1-integrin, and PAI-2 were significantly up-regulated (p < 0.05)in keloid tissue and fibroblasts compared with normal skin. Hsp27, α2ß1-integrin, and PAI-2 expression was inhibited by RNA interference. Both the mRNA and protein levels of Hsp27, α2ß1-integrin, and PAI-2 significantly decreased (p < 0.05) in keloid fibroblasts at 48 hours after transfection. After down-regulation of Hsp27, α2ß1-integrin, and PAI-2, the expression of intracellular extracellular matrix was significantly reduced (p < 0.05). Water-soluble tetrazolium salt-1 assay showed that transfection of Hsp27, α2ß1-integrin, and PAI-2 did not influence the viability/metabolic activity of keloid fibroblasts. CONCLUSIONS: This study demonstrates overexpression of key tension-related proteins in keloid tissue and keloid fibroblasts. Knockdown of Hsp27, PAI-2, and α2ß1-integrin by RNA interference attenuates the expression of mRNA and protein levels and certain other extracellular matrix molecules.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/metabolism , HSP27 Heat-Shock Proteins/physiology , Integrin alpha2beta1/physiology , Keloid/metabolism , Plasminogen Activator Inhibitor 2/physiology , Up-Regulation , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
17.
J Invest Dermatol ; 133(5): 1340-50, 2013 May.
Article in English | MEDLINE | ID: mdl-23303455

ABSTRACT

Mammalian target of rapamycin (mTOR) is essential in controlling several cellular functions. This pathway is dysregulated in keloid disease (KD). KD is a common fibroproliferative dermal lesion with an ill-defined treatment strategy. KD demonstrates excessive matrix deposition, angiogenesis, and inflammatory cell infiltration. In KD, both total and phosphorylated forms of mTOR and p70(S6K)(Thr421/Ser424) are upregulated. Therefore, the aim of this study was to investigate adenosine triphosphate-competitive inhibitors of mTOR kinase previously unreported in keloid and their comparative efficacy with Rapamycin. Here, we present two mTOR kinase inhibitors, KU-0063794 and KU-0068650, that target both mTORC1 and mTORC2 signaling. Treatment with either KU-0063794 or KU-0068650 resulted in complete suppression of Akt, mTORC1, and mTORC2, and inhibition of keloid cell spreading, proliferation, migration, and invasive properties at a very low concentration (2.5 µmol l(-1)). Both KU-0063794 and KU-0068650 significantly (P<0.05) inhibited cell cycle regulation and HIF1-α expression compared with that achieved with Rapamycin alone. In addition, both compounds induced shrinkage and growth arrest in KD, associated with the inhibition of angiogenesis, induction of apoptosis, and reduction in keloid phenotype-associated markers. In contrast, Rapamycin induced minimal antitumor activity. In conclusion, potent dual mTORC1 and mTORC2 inhibitors display therapeutic potential for the treatment of KD.


Subject(s)
Enzyme Inhibitors/therapeutic use , Keloid/drug therapy , Keloid/pathology , Morpholines/therapeutic use , Multiprotein Complexes/antagonists & inhibitors , Pyrimidines/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adolescent , Adult , Aged , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , In Vitro Techniques , Keloid/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Middle Aged , Morpholines/pharmacology , Multiprotein Complexes/drug effects , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Young Adult
18.
Wound Repair Regen ; 21(1): 88-102, 2013.
Article in English | MEDLINE | ID: mdl-23126666

ABSTRACT

Keloid disease (KD) is a fibroproliferative disorder of unknown etiology. Current use of corticosteroid injection is partially beneficial with 80% recurrence rate. Additionally, the efficacy of different steroids, alone or in combination as opposed to monotherapy, in treating KD remains unclear. Here, we compared the single and combined efficacy of glucocorticoids-dexamethasone (Dex), triamcinolone (TAC), and methylprednisolone (Medrol)-on primary keloid fibroblasts (KFs) (n = 27) and normal skin (n = 19) fibroblasts at cellular, protein, and messenger RNA levels in vitro. Our results demonstrated that cytotoxicity to steroids was dose dependent. Cell spreading, attachment, and proliferation were significantly (p < 0.05) reduced by Medrol and TAC. Migration and invasion properties of KF were inhibited significantly (p < 0.05) by Medrol and TAC compared with Dex. At both protein and messenger RNA levels, keloid-associated fibrotic markers were significantly (p < 0.05) decreased by Medrol and TAC compared with Dex. However, vascular endothelial growth factor expression was significantly (p = 0.01) decreased by Dex compared with TAC and Medrol. Medrol and TAC caused significant (p < 0.04) apoptosis, whereas Dex inhibited the UV-induced apoptosis and up-regulated survivin. Blocking of glucocorticoid receptor by RU486 inhibited cytoprotective property of Dex and apoptotic properties of TAC and Medrol. Double treatment with Dex + TAC and Dex + Medrol significantly (p < 0.05) induced apoptosis. In conclusion, this is the first study to report the efficacy of three well-known steroids on KF and suggest that combination may be superior than using a single steroid in treating KD.


Subject(s)
Dexamethasone/pharmacology , Fibroblasts/drug effects , Glucocorticoids/pharmacology , Keloid/drug therapy , Methylprednisolone/pharmacology , Triamcinolone/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Dexamethasone/administration & dosage , Drug Administration Schedule , Fibroblasts/pathology , Glucocorticoids/administration & dosage , Humans , Immunohistochemistry , Injections , Keloid/pathology , Methylprednisolone/administration & dosage , RNA, Messenger/metabolism , Secondary Prevention , Triamcinolone/administration & dosage , Up-Regulation
19.
Wound Repair Regen ; 20(5): 688-706, 2012.
Article in English | MEDLINE | ID: mdl-22985040

ABSTRACT

Keloid disease (KD) is a fibroproliferative disorder of unknown etiopathogenesis with ill-defined treatment. There is increasing evidence to suggest that aberrant Notch signaling may contribute directly to skin pathogenesis and altered expression of Notch receptors identified in KD. Therefore, the aim of this study was to investigate the Notch signaling pathway in KD compared to normal skin (NS). In this study, we employed in vitro primary cell culture models to elucidate the role of Notch signaling in 44 tissue samples from patients with KD split into keloid and extralesional (EL) samples (internal control) from the same patients, and six NS tissue samples (external control). We show the presence of a significant (p < 0.05) up-regulation of Notch receptors and ligand Jagged-1 (JAG-1) in KD compared to EL and NS tissue samples. Cell spreading, attachment, and proliferation were significantly (p < 0.05) reduced in JAG-1 antisense-treated primary dermal fibroblasts isolated from KD and treated with γ-secretase inhibitor (blocks proteolytic cleavage and activation of Notch), evaluated by real-time cell analyzer (RTCA) on a microelectronic sensory array. In contrast, extralesional skin fibroblasts (ELF) treated with recombinant human JAG-1 (rh-JAG-1) peptide showed significant (p < 0.05) enhancement of cell spreading, attachment, and proliferation in RTCA. Activation/inhibition of JAG-1 and Notch signaling significantly (p < 0.05) altered the behavior of primary keloid fibroblasts and ELF, in cell migration (using a scratch wound assay), invasion (using a 3D invasion assay), and angiogenesis (in vitro coculture tube formation assay). In conclusion, this is the first study to demonstrate a potential role for the Notch signaling pathway in KD progression and that targeting this pathway may provide a novel strategy for treatment of KD.


Subject(s)
Calcium-Binding Proteins/metabolism , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Keloid/metabolism , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Skin/metabolism , Wound Healing , Humans , Immunohistochemistry , Jagged-1 Protein , Keloid/pathology , Keloid/physiopathology , Serrate-Jagged Proteins , Signal Transduction , Skin/pathology , Skin/physiopathology , Up-Regulation
20.
Am J Pathol ; 181(5): 1642-58, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22982188

ABSTRACT

Keloid disease (KD) is a fibroproliferative lesion of unknown etiopathogenesis that possibly targets the PI3K/Akt/mTOR pathway. We investigated whether PI3K/Akt/mTOR inhibitor, Palomid 529 (P529), which targets both mammalian target of rapamycin complex 1 (mTORC-1) and mTORC-2 signaling, could exert anti-KD effects in a novel KD organ culture assay and in keloid fibroblasts (KF). Treatment of KF with P529 significantly (P < 0.05) inhibited cell spreading, attachment, proliferation, migration, and invasive properties at a low concentration (5 ng/mL) and induced substantial KF apoptosis when compared with normal dermal fibroblasts. P529 also inhibited hypoxia-inducible factor-1α expression and completely suppressed Akt, GSK3ß, mTOR, eukaryotic initiation factor 4E-binding protein 1, and S6 phosphorylation. P529 significantly (P < 0.05) inhibited proliferating cell nuclear antigen and cyclin D and caused considerable apoptosis. Compared with rapamycin and wortmannin, P529 also significantly (P < 0.05) reduced keloid-associated phenotypic markers in KF. P529 caused tissue shrinkage, growth arrest, and apoptosis in keloid organ cultures and substantially inhibited angiogenesis. pS6, pAkt-Ser473, and mTOR phosphorylation were also suppressed in situ. P529 reduced cellularity and expression of collagen, fibronectin, and α-smooth muscle actin (substantially more than rapamycin). These pre-clinical in vitro and ex vivo observations are evidence that the mTOR pathway is a promising target for future KD therapy and that the dual PI3K/Akt/mTOR inhibitor P529 deserves systematic exploration as a candidate agent for the future treatment of KD.


Subject(s)
Benzopyrans/pharmacology , Keloid/enzymology , Keloid/pathology , Multiprotein Complexes/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adolescent , Adult , Aged , Animals , Apoptosis/drug effects , Benzopyrans/therapeutic use , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Keloid/drug therapy , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Middle Aged , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL