Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Vet Pathol ; : 3009858241252409, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38757523

ABSTRACT

The Cd40l-/- mouse is a well-established model of X-linked hyper-immunoglobulin M (IgM) syndrome, an immunodeficiency disorder of human beings characterized by the lack of expression of the CD40 ligand (CD40L) on activated T-cells, predisposing to infections with opportunistic pathogens like Pneumocystis jirovecii. The aim of our study was to describe the pulmonary lesions in Cd40l-/- mice experimentally infected with Pneumocystis murina, in comparison with naturally infected severe combined immunodeficient (SCID) mice. Formalin-fixed paraffin-embedded lungs from 26 Cd40l-/-, 11 SCID, and 5 uninfected Cd40l-/- mice were examined by histology and immunohistochemistry for the presence of the pathogen and for leukocyte populations (CD3, CD4, CD45R/B220, CD8a, Iba-1, Ly-6G, CD206, MHC II, and NKp46/NCR1). Infection was confirmed by immunohistochemistry in 18/26 (69%) Cd40l-/- mice and in 11/11 (100%) SCID mice. Fourteen out of 26 (54%) Cd40l-/- mice had interstitial pneumonia. Twenty-three out of 26 (88%) Cd40l-/- mice had peribronchiolar/perivascular lymphoplasmacytic infiltrates, rich in B-cells and Mott cells. Acidophilic macrophage pneumonia was additionally found in 20/26 (77%) Cd40l-/- mice. Only 4/11 (36%) SCID mice had interstitial pneumonia, but no peribronchiolar/perivascular infiltrates or acidophilic macrophage pneumonia were observed in this strain. This study represents the first description of pulmonary histopathological lesions in Cd40l-/- mice infected with P. murina. We speculate that the singular characteristics of the inflammatory infiltrates observed in Cd40l-/- mice could be explained by the specific immune phenotype of the model.

3.
Nat Biotechnol ; 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37679541

ABSTRACT

Base and prime editors (BEs and PEs) may provide more precise genetic engineering than nuclease-based approaches because they bypass the dependence on DNA double-strand breaks. However, little is known about their cellular responses and genotoxicity. Here, we compared state-of-the-art BEs and PEs and Cas9 in human hematopoietic stem and progenitor cells with respect to editing efficiency, cytotoxicity, transcriptomic changes and on-target and genome-wide genotoxicity. BEs and PEs induced detrimental transcriptional responses that reduced editing efficiency and hematopoietic repopulation in xenotransplants and also generated DNA double-strand breaks and genotoxic byproducts, including deletions and translocations, at a lower frequency than Cas9. These effects were strongest for cytidine BEs due to suboptimal inhibition of base excision repair and were mitigated by tailoring delivery timing and editor expression through optimized mRNA design. However, BEs altered the mutational landscape of hematopoietic stem and progenitor cells across the genome by increasing the load and relative proportions of nucleotide variants. These findings raise concerns about the genotoxicity of BEs and PEs and warrant further investigation in view of their clinical application.

4.
Mol Ther Methods Clin Dev ; 30: 546-557, 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37693944

ABSTRACT

Hyper-IgM1 is a rare X-linked combined immunodeficiency caused by mutations in the CD40 ligand (CD40LG) gene with a median survival of 25 years, potentially treatable with in situ CD4+ T cell gene editing with Cas9 and a one-size-fits-most corrective donor template. Here, starting from our research-grade editing protocol, we pursued the development of a good manufacturing practice (GMP)-compliant, scalable process that allows for correction, selection and expansion of edited cells, using an integrase defective lentiviral vector as donor template. After systematic optimization of reagents and conditions we proved maintenance of stem and central memory phenotypes and expression and function of CD40LG in edited healthy donor and patient cells recapitulating the physiological CD40LG regulation. We then documented the preserved fitness of edited cells by xenotransplantation into immunodeficient mice. Finally, we transitioned to large-scale manufacturing, and developed a panel of quality control assays. Overall, our GMP-compliant process takes long-range gene editing one step closer to clinical application with a reassuring safety profile.

5.
Blood ; 142(9): 812-826, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37294917

ABSTRACT

Ex vivo gene editing in T cells and hematopoietic stem/progenitor cells (HSPCs) holds promise for treating diseases. Gene editing encompasses the delivery of a programmable editor RNA or ribonucleoprotein, often achieved ex vivo via electroporation, and when aiming for homology-driven correction of a DNA template, often provided by viral vectors together with a nuclease editor. Although HSPCs activate a robust p53-dependent DNA damage response upon nuclease-based editing, the responses triggered in T cells remain poorly characterized. Here, we performed comprehensive multiomics analyses and found that electroporation is the main culprit of cytotoxicity in T cells, causing death and cell cycle delay, perturbing metabolism, and inducing an inflammatory response. Nuclease RNA delivery using lipid nanoparticles (LNPs) nearly abolished cell death and ameliorated cell growth, improving tolerance to the procedure and yielding a higher number of edited cells compared with using electroporation. Transient transcriptomic changes upon LNP treatment were mostly caused by cellular loading with exogenous cholesterol, whose potentially detrimental impact could be overcome by limiting exposure. Notably, LNP-based HSPC editing dampened p53 pathway induction and supported higher clonogenic activity and similar or higher reconstitution by long-term repopulating HSPCs compared with electroporation, reaching comparable editing efficiencies. Overall, LNPs may allow efficient and harmless ex vivo gene editing in hematopoietic cells for the treatment of human diseases.


Subject(s)
Gene Editing , Tumor Suppressor Protein p53 , Humans , Gene Editing/methods , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Hematopoietic Stem Cells/metabolism , RNA/metabolism , CRISPR-Cas Systems
6.
Cell Stem Cell ; 29(10): 1428-1444.e9, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36206730

ABSTRACT

Long-range gene editing by homology-directed repair (HDR) in hematopoietic stem/progenitor cells (HSPCs) often relies on viral transduction with recombinant adeno-associated viral vector (AAV) for template delivery. Here, we uncover unexpected load and prolonged persistence of AAV genomes and their fragments, which trigger sustained p53-mediated DNA damage response (DDR) upon recruiting the MRE11-RAD50-NBS1 (MRN) complex on the AAV inverted terminal repeats (ITRs). Accrual of viral DNA in cell-cycle-arrested HSPCs led to its frequent integration, predominantly in the form of transcriptionally competent ITRs, at nuclease on- and off-target sites. Optimized delivery of integrase-defective lentiviral vector (IDLV) induced lower DNA load and less persistent DDR, improving clonogenic capacity and editing efficiency in long-term repopulating HSPCs. Because insertions of viral DNA fragments are less frequent with IDLV, its choice for template delivery mitigates the adverse impact and genotoxic burden of HDR editing and should facilitate its clinical translation in HSPC gene therapy.


Subject(s)
DNA, Viral , Tumor Suppressor Protein p53 , CRISPR-Cas Systems , DNA Damage , Gene Editing , Hematopoietic Stem Cells , Humans , Integrases , Tumor Suppressor Protein p53/genetics
7.
Cell ; 185(13): 2248-2264.e21, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35617958

ABSTRACT

Hematopoietic stem/progenitor cell gene therapy (HSPC-GT) is proving successful to treat several genetic diseases. HSPCs are mobilized, harvested, genetically corrected ex vivo, and infused, after the administration of toxic myeloablative conditioning to deplete the bone marrow (BM) for the modified cells. We show that mobilizers create an opportunity for seamless engraftment of exogenous cells, which effectively outcompete those mobilized, to repopulate the depleted BM. The competitive advantage results from the rescue during ex vivo culture of a detrimental impact of mobilization on HSPCs and can be further enhanced by the transient overexpression of engraftment effectors exploiting optimized mRNA-based delivery. We show the therapeutic efficacy in a mouse model of hyper IgM syndrome and further developed it in human hematochimeric mice, showing its applicability and versatility when coupled with gene transfer and editing strategies. Overall, our findings provide a potentially valuable strategy paving the way to broader and safer use of HSPC-GT.


Subject(s)
Gene Editing , Hematopoietic Stem Cell Transplantation , Animals , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells , Humans , Mice
8.
Nat Protoc ; 16(6): 2991-3025, 2021 06.
Article in English | MEDLINE | ID: mdl-34031609

ABSTRACT

Gene editing by engineered nucleases has revolutionized the field of gene therapy by enabling targeted and precise modification of the genome. However, the limited availability of methods for clonal tracking of edited cells has resulted in a paucity of information on the diversity, abundance and behavior of engineered clones. Here we detail the wet laboratory and bioinformatic BAR-Seq pipeline, a strategy for clonal tracking of cells harboring homology-directed targeted integration of a barcoding cassette. We present the BAR-Seq web application, an online, freely available and easy-to-use software that allows performing clonal tracking analyses on raw sequencing data without any computational resources or advanced bioinformatic skills. BAR-Seq can be applied to most editing strategies, and we describe its use to investigate the clonal dynamics of human edited hematopoietic stem/progenitor cells in xenotransplanted hosts. Notably, BAR-Seq may be applied in both basic and translational research contexts to investigate the biology of edited cells and stringently compare editing protocols at a clonal level. Our BAR-Seq pipeline allows library preparation and validation in a few days and clonal analyses of edited cell populations in 1 week.


Subject(s)
Cell Tracking/methods , Clone Cells , Gene Editing , Software , DNA Barcoding, Taxonomic
9.
EMBO Mol Med ; 13(3): e13545, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33475257

ABSTRACT

Precise correction of the CD40LG gene in T cells and hematopoietic stem/progenitor cells (HSPC) holds promise for treating X-linked hyper-IgM Syndrome (HIGM1), but its actual therapeutic potential remains elusive. Here, we developed a one-size-fits-all editing strategy for effective T-cell correction, selection, and depletion and investigated the therapeutic potential of T-cell and HSPC therapies in the HIGM1 mouse model. Edited patients' derived CD4 T cells restored physiologically regulated CD40L expression and contact-dependent B-cell helper function. Adoptive transfer of wild-type T cells into conditioned HIGM1 mice rescued antigen-specific IgG responses and protected mice from a disease-relevant pathogen. We then obtained ~ 25% CD40LG editing in long-term repopulating human HSPC. Transplanting such proportion of wild-type HSPC in HIGM1 mice rescued immune functions similarly to T-cell therapy. Overall, our findings suggest that autologous edited T cells can provide immediate and substantial benefits to HIGM1 patients and position T-cell ahead of HSPC gene therapy because of easier translation, lower safety concerns and potentially comparable clinical benefits.


Subject(s)
Hyper-IgM Immunodeficiency Syndrome, Type 1 , Hyper-IgM Immunodeficiency Syndrome , Animals , Gene Editing , Hematopoietic Stem Cells , Humans , Mice , T-Lymphocytes
10.
Nat Biotechnol ; 38(11): 1298-1308, 2020 11.
Article in English | MEDLINE | ID: mdl-32601433

ABSTRACT

Targeted gene editing in hematopoietic stem cells (HSCs) is a promising treatment for several diseases. However, the limited efficiency of homology-directed repair (HDR) in HSCs and the unknown impact of the procedure on clonal composition and dynamics of transplantation have hampered clinical translation. Here, we apply a barcoding strategy to clonal tracking of edited cells (BAR-Seq) and show that editing activates p53, which substantially shrinks the HSC clonal repertoire in hematochimeric mice, although engrafted edited clones preserve multilineage and self-renewing capacity. Transient p53 inhibition restored polyclonal graft composition. We increased HDR efficiency by forcing cell-cycle progression and upregulating components of the HDR machinery through transient expression of the adenovirus 5 E4orf6/7 protein, which recruits the cell-cycle controller E2F on its target genes. Combined E4orf6/7 expression and p53 inhibition resulted in HDR editing efficiencies of up to 50% in the long-term human graft, without perturbing repopulation and self-renewal of edited HSCs. This enhanced protocol should broaden applicability of HSC gene editing and pave its way to clinical translation.


Subject(s)
Cell Tracking , Gene Editing , Hematopoietic Stem Cells/cytology , Animals , Base Sequence , Cell Lineage , Clone Cells , Dependovirus/metabolism , G2 Phase , HEK293 Cells , Humans , Mice , Recombinational DNA Repair , Reproducibility of Results , S Phase , Transcription, Genetic , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Viral Proteins/metabolism , Xenograft Model Antitumor Assays
11.
Cell Stem Cell ; 24(4): 551-565.e8, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905619

ABSTRACT

Precise gene editing in hematopoietic stem and progenitor cells (HSPCs) holds promise for treating genetic diseases. However, responses triggered by programmable nucleases in HSPCs are poorly characterized and may negatively impact HSPC engraftment and long-term repopulation capacity. Here, we induced either one or several DNA double-stranded breaks (DSBs) with optimized zinc-finger and CRISPR/Cas9 nucleases and monitored DNA damage response (DDR) foci induction, cell-cycle progression, and transcriptional responses in HSPC subpopulations, with up to single-cell resolution. p53-mediated DDR pathway activation was the predominant response to even single-nuclease-induced DSBs across all HSPC subtypes analyzed. Excess DSB load and/or adeno-associated virus (AAV)-mediated delivery of DNA repair templates induced cumulative p53 pathway activation, constraining proliferation, yield, and engraftment of edited HSPCs. However, functional impairment was reversible when DDR burden was low and could be overcome by transient p53 inhibition. These findings provide molecular and functional evidence for feasible and seamless gene editing in HSPCs.


Subject(s)
DNA Damage , Gene Editing , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Humans , K562 Cells , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
12.
Sci Transl Med ; 9(411)2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29021165

ABSTRACT

Targeted genome editing in hematopoietic stem/progenitor cells (HSPCs) is an attractive strategy for treating immunohematological diseases. However, the limited efficiency of homology-directed editing in primitive HSPCs constrains the yield of corrected cells and might affect the feasibility and safety of clinical translation. These concerns need to be addressed in stringent preclinical models and overcome by developing more efficient editing methods. We generated a humanized X-linked severe combined immunodeficiency (SCID-X1) mouse model and evaluated the efficacy and safety of hematopoietic reconstitution from limited input of functional HSPCs, establishing thresholds for full correction upon different types of conditioning. Unexpectedly, conditioning before HSPC infusion was required to protect the mice from lymphoma developing when transplanting small numbers of progenitors. We then designed a one-size-fits-all IL2RG (interleukin-2 receptor common γ-chain) gene correction strategy and, using the same reagents suitable for correction of human HSPC, validated the edited human gene in the disease model in vivo, providing evidence of targeted gene editing in mouse HSPCs and demonstrating the functionality of the IL2RG-edited lymphoid progeny. Finally, we optimized editing reagents and protocol for human HSPCs and attained the threshold of IL2RG editing in long-term repopulating cells predicted to safely rescue the disease, using clinically relevant HSPC sources and highly specific zinc finger nucleases or CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein 9). Overall, our work establishes the rationale and guiding principles for clinical translation of SCID-X1 gene editing and provides a framework for developing gene correction for other diseases.


Subject(s)
Hematopoietic Stem Cells/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Gene Targeting/methods , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Mice , Mice, SCID
13.
Tumori ; 102(2): 144-9, 2016.
Article in English | MEDLINE | ID: mdl-26917410

ABSTRACT

PURPOSE: Because the peptidyl-prolyl isomerase PIN1 interacts with multiple protein kinases and phosphoproteins into a network orchestrating the cellular response to various stimuli, there is an increasing interest in exploiting its potential as therapeutic target. In the present study, the effect of targeting PIN1 was investigated in 2 human cancer cell lines characterized by increased aggressive potential, high expression of erbB receptor family members, and defective p53. METHODS: PIN1 silencing was carried out in skin squamous cell carcinoma A431 cells displaying elevated EGFR/HER1 levels and in ovarian adenocarcinoma SKOV-3 cells displaying high levels of erbB2 (HER2). Nonoverlapping siRNA duplexes targeting different regions of PIN1 mRNA were transfected in tumor cells, which were analyzed using Western blotting for the expression of selected proteins. In vivo tumorigenicity studies were carried out in athymic nude mice. RESULTS: A431 and SKOV-3 cell systems were found to be a source of cells with increased aggressive potential, i.e., cancer stem cell-like cells, as defined by the capability to grow as spheres. A marked decrease of PIN1 levels and of sphere-forming capability was observed in PIN1-silenced cells. The expression of phospho-p38 decreased following PIN1 silencing in A431 and SKOV-3 cells, as well as phospho-EGFR levels in A431 - silenced cells. PIN1 inhibition prolonged latency and reduced tumor take and growth of SKOV-3 cells in nude mice. CONCLUSIONS: Our results support that PIN1 may be a valuable target to hit in cancer cells characterized by increased aggressive potential, overexpression of erbB receptor family members, and defective p53.


Subject(s)
ErbB Receptors/metabolism , Gene Silencing , Neoplasms/metabolism , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/metabolism , Animals , Blotting, Western , Carcinogenicity Tests , Carcinoma, Squamous Cell/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase , Ovarian Neoplasms/metabolism , Skin Neoplasms/metabolism , Up-Regulation
14.
Proc Natl Acad Sci U S A ; 110(10): 3931-6, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23417300

ABSTRACT

DNA damage leads to a halt in proliferation owing to apoptosis or senescence, which prevents transmission of DNA alterations. This cellular response depends on the tumor suppressor p53 and functions as a powerful barrier to tumor development. Adult stem cells are resistant to DNA damage-induced apoptosis or senescence, however, and how they execute this response and suppress tumorigenesis is unknown. We show that irradiation of hematopoietic and mammary stem cells up-regulates the cell cycle inhibitor p21, a known target of p53, which prevents p53 activation and inhibits p53 basal activity, impeding apoptosis and leading to cell cycle entry and symmetric self-renewing divisions. p21 also activates DNA repair, limiting DNA damage accumulation and self-renewal exhaustion. Stem cells with moderate DNA damage and diminished self-renewal persist after irradiation, however. These findings suggest that stem cells have evolved a unique, p21-dependent response to DNA damage that leads to their immediate expansion and limits their long-term survival.


Subject(s)
Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Adult Stem Cells/radiation effects , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Cell Cycle Checkpoints/physiology , Cell Cycle Checkpoints/radiation effects , Cell Division/radiation effects , DNA Repair , Female , Hematopoietic Stem Cells/radiation effects , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/radiation effects , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Up-Regulation/radiation effects
15.
J Cell Physiol ; 228(4): 871-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23018838

ABSTRACT

Many squamous cell carcinomas (SCCs) are characterized by high levels of EGFR and by overexpression of the ΔNp63α isoform. Here, we investigated the regulation of ΔNp63α expression upon EGFR activation and the role of the EGFR-ΔNp63α axis in proliferation of SCC tumor-initiating cells (TICs). SCC cell lines A-431, Cal-27, and SCC-25 treated with EGF showed a time-dependent increase in ΔNp63α expression at the protein and mRNA levels, which was blocked by the tyrosine kinase inhibitor (TKI) Lapatinib. RNA interference experiments suggested the role of STAT3 in regulating ΔNp63α expression downstream of EGFR. Inactivation of EGFR by the monoclonal antibody Cetuximab and RNA interference against STAT3 or ΔNp63α impaired the TICs ability to grow under non-differentiating conditions. Radiation treatment, which triggers EGFR activation, induced ΔNp63α accumulation without affecting TICs proliferation, whereas the combination Cetuximab plus radiation significantly reduced TICs growth under non-differentiating conditions. Together, our findings provide evidence that ΔNp63α expression is regulated by EGFR activation through STAT3 and that the EGFR-ΔNp63α axis is crucial for proliferation of TICs present in SCCs.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , ErbB Receptors/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , STAT3 Transcription Factor/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Carcinoma, Squamous Cell/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , ErbB Receptors/genetics , Humans , Lapatinib , MCF-7 Cells , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , RNA, Messenger/genetics , STAT3 Transcription Factor/genetics
16.
Cancer Lett ; 296(2): 206-15, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20435407

ABSTRACT

There is indication that tumor growth is sustained by subpopulation of cells with stem-like features but little is known on their genomic characterization and their genetic stability. We report a detailed molecular cytogenetic characterization using Spectral Karyotyping and fluorescent in situ hybridization of parental serum-cultured adherent cells and their sphere-growing stem-like counterpart before and after differentiation from six cell lines established from solid tumors. Our findings indicate increased cytogenetic complexity in sphere-growing stem-like and their differentiated adherent cells compared to parental adherent component suggesting the existence within cell lines of heterogeneous and genetically unstable subpopulations of cells endowed with stem-like features.


Subject(s)
Cell Line, Tumor/pathology , Stem Cells/pathology , Antigens, CD/analysis , Biomarkers, Tumor/analysis , Cell Adhesion , Cell Culture Techniques/methods , Cell Differentiation , Cell Division , Chromosome Aberrations , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Karyotyping/methods , Neoplasms/genetics
17.
Clin Cancer Res ; 15(6): 2010-21, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276287

ABSTRACT

PURPOSE: The existence of tumor-initiating cells in breast cancer has profound implications for cancer therapy. In this study, we investigated the sensitivity of tumor-initiating cells isolated from human epidermal growth factor receptor type 2 (HER2)-overexpressing carcinoma cell lines to trastuzumab, a compound used for the targeted therapy of breast cancer. EXPERIMENTAL DESIGN: Spheres were analyzed by indirect immunofluorescence for HER2 cell surface expression and by real-time PCR for HER2 mRNA expression in the presence or absence of the Notch1 signaling inhibitor (GSI) or Notch1 small interfering RNA. Xenografts of HER2-overexpressing breast tumor cells were treated with trastuzumab or doxorubicin. The sphere-forming efficiency (SFE) and serial transplantability of tumors were assessed. RESULTS: In HER2-overexpressing carcinoma cell lines, cells with tumor-initiating cell properties presented increased HER2 levels compared with the bulk cell population without modification in HER2 gene amplification. HER2 levels were controlled by Notch1 signaling, as shown by the reduction of HER2 cell surface expression and lower SFE following gamma-secretase inhibition or Notch1 specific silencing. We also show that trastuzumab was able to effectively target tumor-initiating cells of HER2-positive carcinoma cell lines, as indicated by the significant decrease in SFE and the loss of serial transplantability, following treatment of HER2-overexpressing xenotransplants. CONCLUSIONS: Here, we provide evidence for the therapeutic efficacy of trastuzumab in debulking and in targeting tumor-initiating cells of HER2-overexpressing tumors. We also propose that Notch signaling regulates HER2 expression, thereby representing a critical survival pathway of tumor-initiating cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/analysis , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Humans , Lapatinib , Mice , Quinazolines/pharmacology , RNA, Messenger/analysis , Receptor, ErbB-2/genetics , Receptor, Notch1/physiology , Trastuzumab , Xenograft Model Antitumor Assays
18.
Cancer Res ; 67(11): 5308-17, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545611

ABSTRACT

In some HER2-positive breast tumors, cell surface overexpression of HER2 is not associated with gene amplification but may instead rest in altered gene transcription, half-life, or recycling of the oncoprotein. Here, we show that HER2 overexpression in HER2 2+ carcinomas is associated with neither an increase in gene transcription nor a deregulation in the ubiquitin-dependent pathways, but instead seems to be regulated by protein kinase Calpha (PKCalpha) activity. The stimulation of PKCalpha up-regulated HER2 expression, whereas PKCalpha inhibition by pharmacologic treatments and PKCalpha-specific small interfering RNA led to a dramatic down-regulation of HER2 levels only in breast cancer cells HER2 2+. Consistent with the in vitro data, our biochemical analysis of HER2 2+ human primary breast specimens revealed significantly higher levels of phosphorylated PKCalpha compared with HER2-negative tumors. Inhibition of HER2 activation by the tyrosine kinase inhibitor lapatinib led to decreased levels of PKCalpha phosphorylation, clearly indicating a cross-talk between PKCalpha and HER2 molecules. These data suggest that HER2 overexpression in HER2 2+ carcinomas is due to an accumulation of the recycled oncoprotein to the cell surface induced by activated PKCalpha.


Subject(s)
Breast Neoplasms/enzymology , Protein Kinase C-alpha/metabolism , Receptor, ErbB-2/biosynthesis , Breast Neoplasms/genetics , Carbazoles/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Gene Amplification , Humans , Immunoprecipitation , Indoles/pharmacology , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/biosynthesis , Protein Kinase C-alpha/genetics , Proto-Oncogene Proteins c-cbl/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...