Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Int J Mol Sci ; 24(10)2023 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-37240396

RESUMO

The main aim of this study is to synthesize contrast microbubbles (MB) functionalized with engineered protein ligands using a microfluidic device to target breast cancer specific vascular B7-H3 receptor in vivo for diagnostic ultrasound imaging. We used a high-affinity affibody (ABY) selected against human/mouse B7-H3 receptor for engineering targeted MBs (TMBs). We introduced a C-terminal cysteine residue to this ABY ligand for facilitating site-specific conjugation to DSPE-PEG-2K-maleimide (M. Wt = 2.9416 kDa) phospholipid for MB formulation. We optimized the reaction conditions of bioconjugations and applied it for microfluidic based synthesis of TMBs using DSPE-PEG-ABY and DPPC liposomes (5:95 mole %). The binding affinity of TMBs to B7-H3 (MBB7-H3) was tested in vitro in MS1 endothelial cells expressing human B7-H3 (MS1B7-H3) by flow chamber assay, and by ex vivo in the mammary tumors of a transgenic mouse model (FVB/N-Tg (MMTV-PyMT)634Mul/J), expressing murine B7-H3 in the vascular endothelial cells by immunostaining analyses. We successfully optimized the conditions needed for generating TMBs using a microfluidic system. The synthesized MBs showed higher affinity to MS1 cells engineered to express higher level of hB7-H3, and in the endothelial cells of mouse tumor tissue upon injecting TMBs in a live animal. The average number (mean ± SD) of MBB7-H3 binding to MS1B7-H3 cells was estimated to be 354.4 ± 52.3 per field of view (FOV) compared to wild-type control cells (MS1WT; 36.2 ± 7.5/FOV). The non-targeted MBs did not show any selective binding affinity to both the cells (37.7 ± 7.8/FOV for MS1B7-H3 and 28.3 ± 6.7/FOV for MS1WT cells). The fluorescently labeled MBB7-H3 upon systemic injection in vivo co-localized to tumor vessels, expressing B7-H3 receptor, as validated by ex vivo immunofluorescence analyses. We have successfully synthesized a novel MBB7-H3 via microfluidic device, which allows us to produce on demand TMBs for clinical applications. This clinically translatable MBB7-H3 showed significant binding affinity to vascular endothelial cells expressing B7-H3 both in vitro and in vivo, which shows its potential for clinical translation as a molecular ultrasound contrast agent for human applications.


Assuntos
Neoplasias da Mama , Receptores Histamínicos H3 , Camundongos , Animais , Humanos , Feminino , Microbolhas , Células Endoteliais/metabolismo , Ultrassonografia/métodos , Camundongos Transgênicos , Imagem Molecular/métodos , Meios de Contraste , Neoplasias da Mama/patologia , Dispositivos Lab-On-A-Chip
2.
Bioconjug Chem ; 30(6): 1677-1689, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31082216

RESUMO

Spectroscopic photoacoustic (sPA) molecular imaging has high potential for identification of exogenous contrast agents targeted to specific markers. Antibody-dye conjugates have recently been used extensively for preclinical sPA and other optical imaging modalities for highly specific molecular imaging of breast cancer. However, antibody-based agents suffer from long circulation times that limit image specificity. Here, the efficacy of a small protein scaffold, the affibody (ABY), conjugated to indocyanine green (ICG), a near-infrared fluorescence dye, as a targeted molecular imaging probe is demonstrated. In particular, B7-H3 (CD276), a cellular receptor expressed in breast cancer, was imaged via sPA and fluorescence molecular imaging to differentiate invasive tumors from normal glands in mice. Administration of ICG conjugated to an ABY specific to B7-H3 (ABYB7-H3-ICG) showed significantly higher signal in mammary tumors compared to normal glands of mice. ABYB7-H3-ICG is a compelling scaffold for molecular sPA imaging for breast cancer detection.


Assuntos
Antígenos B7/análise , Neoplasias da Mama/diagnóstico por imagem , Meios de Contraste/química , Corantes Fluorescentes/química , Imunoconjugados/química , Verde de Indocianina/química , Animais , Feminino , Camundongos , Imagem Óptica/métodos , Técnicas Fotoacústicas/métodos
3.
Wound Repair Regen ; 25(6): 964-971, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29316036

RESUMO

Wound healing is characterized by the production of large amounts of protein necessary to replace lost cellular mass and extracellular matrix. The unfolded protein response (UPR) is an important adaptive cellular response to increased protein synthesis. One of the main components of the UPR is IRE1, an endoplasmic reticulum transmembrane protein with endonuclease activity that produces the activated form of the transcription factor XBP1. Using luciferase reporter mice for Xbp1 splicing, we showed that IRE1 was up-regulated during excisional wound healing at the time in wound healing consistent with that of the proliferative phase, when the majority of protein synthesis for cellular proliferation and matrix deposition occurs. Furthermore, using a small molecule inhibitor of IRE1 we demonstrated that inhibition of IRE1 led to decreased scar formation in treated mice. Results were recapitulated in a hypertrophic scar mouse model. These data help provide a cellular pathway to target in the treatment of hypertrophic scarring and keloid disorders.


Assuntos
Cicatriz Hipertrófica/metabolismo , Cicatriz/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Animais , Proliferação de Células , Matriz Extracelular/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Resposta a Proteínas não Dobradas , Regulação para Cima , Cicatrização
4.
Blood ; 124(13): 2051-60, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25061178

RESUMO

Secreted protein CCN1, encoded by CYR61, is involved in wound healing, angiogenesis, and osteoblast differentiation. We identified CCN1 as a microenvironmental factor produced by mesenchymal cells and overexpressed in bones of a subset of patients with monoclonal gammopathy of undetermined significance (MGUS), asymptomatic myeloma (AMM), and multiple myeloma (MM). Our analysis showed that overexpression of CYR61 was independently associated with superior overall survival of MM patients enrolled in our Total Therapy 3 protocol. Moreover, elevated CCN1 was associated with a longer time for MGUS/AMM to progress to overt MM. During remission from MM, high levels of CCN1 were associated with superior progression-free and overall survival and stratified patients with molecularly defined high-risk MM. Recombinant CCN1 directly inhibited in vitro growth of MM cells, and overexpression of CYR61 in MM cells reduced tumor growth and prevented bone destruction in vivo in severe combined immunodeficiency-hu mice. Signaling through αvß3 was required for CCN1 prevention of bone disease. CYR61 expression may signify early perturbation of the microenvironment before conversion to overt MM and may be a compensatory mechanism to control MM progression. Therapeutics that upregulate CYR61 should be investigated for treating MM bone disease.


Assuntos
Doenças Ósseas/etiologia , Proteína Rica em Cisteína 61/genética , Expressão Gênica , Mieloma Múltiplo/complicações , Mieloma Múltiplo/genética , Microambiente Tumoral/genética , Animais , Doenças Assintomáticas , Biópsia , Medula Óssea/metabolismo , Medula Óssea/patologia , Osso e Ossos/patologia , Linhagem Celular Tumoral , Proteína Rica em Cisteína 61/sangue , Proteína Rica em Cisteína 61/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Xenoenxertos , Humanos , Camundongos , Gamopatia Monoclonal de Significância Indeterminada/genética , Gamopatia Monoclonal de Significância Indeterminada/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/mortalidade , Prognóstico
5.
BMC Cancer ; 15: 864, 2015 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-26545722

RESUMO

BACKGROUND: Human primary myeloma (MM) cells do not survive in culture; current in vitro and in vivo systems for growing these cells are limited to coculture with a specific bone marrow (BM) cell type or growth in an immunodeficient animal model. The purpose of the study is to establish an interactive healthy donor whole BM based culture system capable of maintaining prolonged survival of primary MM cells. This normal BM (NBM) coculture system is different from using autologous BM that is already affected by the disease. METHODS: Whole BM from healthy donors was cultured in medium supplemented with BM serum from MM patients for 7 days, followed by 7 days of coculture with CD138-selected primary MM cells or MM cell lines. MM cells in the coculture were quantified using flow cytometry or bioluminescence of luciferase-expressing MM cells. T-cell cytokine array and proteomics were performed to identify secreted factors. RESULTS: NBM is composed of adherent and nonadherent compartments containing typical hematopoietic and mesenchymal cells. MM cells, or a subset of MM cells, from all examined cases survived and grew in this system, regardless of the MM cells' molecular risk or subtype, and growth was comparable to coculture with individual stromal cell types. Adherent and nonadherent compartments supported MM growth, and this support required patient serum for optimal growth. Increased levels of MM growth factors IL-6 and IL-10 along with MM clinical markers B2M and LDHA were detected in supernatants from the NBM coculture than from the BM cultured alone. Levels of extracellular matrix factors (e.g., MMP1, HMCN1, COL3A1, ACAN) and immunomodulatory factors (e.g., IFI16, LILRB4, PTPN6, AZGP1) were changed in the coculture system. The NBM system protected MM cells from dexamethasone but not bortezomib, and effects of lenalidomide varied. CONCLUSIONS: The NBM system demonstrates the ability of primary MM plasma cells to interact with and to survive in coculture with healthy adult BM. This model is suitable for studying MM-microenvironment interactions, particularly at the early stage of engagement in new BM niches, and for characterizing MM cell subpopulations capable of long-term survival through secretion of extracellular matrix and immune-related factors.


Assuntos
Células da Medula Óssea/metabolismo , Comunicação Celular , Técnicas de Cocultura , Mieloma Múltiplo/metabolismo , Antígenos CD/metabolismo , Antineoplásicos/farmacologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular , Citocinas/biossíntese , Humanos , Imuno-Histoquímica , Imunofenotipagem , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Mieloma Múltiplo/patologia , Osteoclastos/metabolismo , Proteoma , Proteômica/métodos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Microambiente Tumoral
6.
Nat Commun ; 15(1): 4114, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38750057

RESUMO

Cellular sensitivity to ferroptosis is primarily regulated by mechanisms mediating lipid hydroperoxide detoxification. We show that inositol-requiring enzyme 1 (IRE1α), an endoplasmic reticulum (ER) resident protein critical for the unfolded protein response (UPR), also determines cellular sensitivity to ferroptosis. Cancer and normal cells depleted of IRE1α gain resistance to ferroptosis, while enhanced IRE1α expression promotes sensitivity to ferroptosis. Mechanistically, IRE1α's endoribonuclease activity cleaves and down-regulates the mRNA of key glutathione biosynthesis regulators glutamate-cysteine ligase catalytic subunit (GCLC) and solute carrier family 7 member 11 (SLC7A11). This activity of IRE1α is independent of its role in regulating the UPR and is evolutionarily conserved. Genetic deficiency and pharmacological inhibition of IRE1α have similar effects in inhibiting ferroptosis and reducing renal ischemia-reperfusion injury in mice. Our findings reveal a previously unidentified role of IRE1α to regulate ferroptosis and suggests inhibition of IRE1α as a promising therapeutic strategy to mitigate ferroptosis-associated pathological conditions.


Assuntos
Sistema y+ de Transporte de Aminoácidos , Endorribonucleases , Ferroptose , Glutationa , Proteínas Serina-Treonina Quinases , Ferroptose/genética , Endorribonucleases/metabolismo , Endorribonucleases/genética , Animais , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Camundongos , Glutationa/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Glutamato-Cisteína Ligase/metabolismo , Glutamato-Cisteína Ligase/genética , Resposta a Proteínas não Dobradas , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/genética , Linhagem Celular Tumoral , Camundongos Endogâmicos C57BL , Masculino , Camundongos Knockout
7.
Am J Hematol ; 88(6): 463-71, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23456977

RESUMO

Myeloma cells typically grow in bone, recruit osteoclast precursors and induce their differentiation and activity in areas adjacent to tumor foci. Bruton's tyrosine kinase (BTK), of the TEC family, is expressed in hematopoietic cells and is particularly involved in B-lymphocyte function and osteoclastogenesis. We demonstrated BTK expression in clinical myeloma plasma cells, interleukin (IL)-6- or stroma-dependent cell lines and osteoclasts. SDF-1 induced BTK activation in myeloma cells and BTK inhibition by small hairpin RNA or the small molecule inhibitor, LFM-A13, reduced their migration toward stromal cell-derived factor-1 (SDF-1). Pretreatment with LFM-A13 also reduced in vivo homing of myeloma cells to bone using bioluminescence imaging in the SCID-rab model. Enforced expression of BTK in myeloma cell line enhanced cell migration toward SDF-1 but had no effect on short-term growth. BTK expression was correlated with cell-surface CXCR4 expression in myeloma cells (n = 33, r = 0.81, P < 0.0001), and BTK gene and protein expression was more profound in cell-surface CXCR4-expressing myeloma cells. BTK was not upregulated by IL-6 while its inhibition had no effect on IL-6 signaling in myeloma cells. Human osteoclast precursors also expressed BTK and cell-surface CXCR4 and migrated toward SDF-1. LFM-A13 suppressed migration and differentiation of osteoclast precursors as well as bone-resorbing activity of mature osteoclasts. In primary myeloma-bearing SCID-rab mice, LFM-A13 inhibited osteoclast activity, prevented myeloma-induced bone resorption and moderately suppressed myeloma growth. These data demonstrate BTK and cell-surface CXCR4 association in myeloma cells and that BTK plays a role in myeloma cell homing to bone and myeloma-induced bone disease. Am. J. Hematol. 88:463-471, 2013. © 2013 Wiley Periodicals, Inc.


Assuntos
Doenças Ósseas/enzimologia , Doenças Ósseas/patologia , Movimento Celular/fisiologia , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Proteínas Tirosina Quinases/metabolismo , Tirosina Quinase da Agamaglobulinemia , Amidas/farmacologia , Animais , Doenças Ósseas/prevenção & controle , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Quimiocina CXCL12/metabolismo , Humanos , Camundongos , Camundongos SCID , Nitrilas/farmacologia , Osteoclastos/enzimologia , Osteoclastos/patologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Receptores CXCR4/metabolismo , Transdução de Sinais
8.
Sci Rep ; 11(1): 23026, 2021 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-34845270

RESUMO

Molecular imaging using singlechain variable fragments (scFv) of antibodies targeting cancer specific antigens have been considered a non-immunogenic approach for early diagnosis in the clinic. Usually, production of proteins is performed within Escherichia coli. Recombinant proteins are either expressed in E. coli cytoplasm as insoluble inclusion bodies, that often need cumbersome denaturation and refolding processes, or secreted toward the periplasm as soluble proteins that highly reduce the overall yield. However, production of active scFvs in their native form, without any heterologous fusion, is required for clinical applications. In this study, we expressed an anti-thymocyte differentiation antigen-scFv (Thy1-scFv) as a fusion protein with a N-terminal sequence including 3 × hexa-histidines, as purification tags, together with a Trx-tag and a S-tag for enhanced-solubility. Our strategy allowed to recover ~ 35% of Thy1-scFv in the soluble cytoplasmic fraction. An enterokinase cleavage site in between Thy1-scFv and the upstream tags was used to regenerate the protein with 97.7 ± 2.3% purity without any tags. Thy1-scFv showed functionality towards its target on flow cytometry assays. Finally, in vivo molecular imaging using Thy1-scFv conjugated to an ultrasound contrast agent (MBThy1-scFv) demonstrated signal enhancement on a transgenic pancreatic ductal adenocarcinoma (PDAC) mouse model (3.1 ± 1.2 a.u.) compared to non-targeted control (0.4 ± 0.4 a.u.) suggesting potential for PDAC early diagnosis. Overall, our strategy facilitates the expression and purification of Thy1-scFv while introducing its ability for diagnostic molecular imaging of pancreatic cancer. The presented methodology could be expanded to other important eukaryotic proteins for various applications, including but not limited to molecular imaging.


Assuntos
Imagem Molecular/instrumentação , Anticorpos de Cadeia Única/imunologia , Animais , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/terapia , Cromatografia de Afinidade , Meios de Contraste/química , Citoplasma/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Escherichia coli/metabolismo , Citometria de Fluxo , Vetores Genéticos , Histidina/química , Humanos , Corpos de Inclusão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Imagem Molecular/métodos , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/terapia , Periplasma/metabolismo , Proteínas Recombinantes/química , Timócitos/citologia , Pesquisa Translacional Biomédica
9.
Bioeng Transl Med ; 6(1): e10183, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33532585

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is often associated with a poor prognosis due to silent onset, resistance to therapies, and rapid spreading. Most patients are ineligible for curable surgery as they present with advanced disease at the time of diagnosis. Present diagnostic methods relying on anatomical changes have various limitations including difficulty to discriminate between benign and malignant conditions, invasiveness, the ambiguity of imaging results, or the inability to detect molecular biomarkers of PDAC initiation and progression. Therefore, new imaging technologies with high sensitivity and specificity are critically needed for accurately detecting PDAC and noninvasively characterizing molecular features driving its pathogenesis. Contrast enhanced targeted ultrasound (CETUS) is an upcoming molecular imaging modality that specifically addresses these issues. Unlike anatomical imaging modalities such as CT and MRI, molecular imaging using CETUS is promising for early and accurate detection of PDAC. The use of molecularly targeted microbubbles that bind to neovascular targets can enhance the ultrasound signal specifically from malignant PDAC tissues. This review discusses the current state of diagnostic imaging modalities for pancreatic cancer and places a special focus on ultrasound targeted-microbubble technology together with its clinical translatability for PDAC detection.

10.
IEEE Trans Med Imaging ; 39(10): 3079-3088, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32286963

RESUMO

Ultrasound molecular imaging (UMI) is enabled by targeted microbubbles (MBs), which are highly reflective ultrasound contrast agents that bind to specific biomarkers. Distinguishing between adherent MBs and background signals can be challenging in vivo. The preferred preclinical technique is differential targeted enhancement (DTE), wherein a strong acoustic pulse is used to destroy MBs to verify their locations. However, DTE intrinsically cannot be used for real-time imaging and may cause undesirable bioeffects. In this work, we propose a simple 4-layer convolutional neural network to nondestructively detect adherent MB signatures. We investigated several types of input data to the network: "anatomy-mode" (fundamental frequency), "contrast-mode" (pulse-inversion harmonic frequency), or both, i.e., "dual-mode", using IQ channel signals, the channel sum, or the channel sum magnitude. Training and evaluation were performed on in vivo mouse tumor data and microvessel phantoms. The dual-mode channel signals yielded optimal performance, achieving a soft Dice coefficient of 0.45 and AUC of 0.91 in two test images. In a volumetric acquisition, the network best detected a breast cancer tumor, resulting in a generalized contrast-to-noise ratio (GCNR) of 0.93 and Kolmogorov-Smirnov statistic (KSS) of 0.86, outperforming both regular contrast mode imaging (GCNR = 0.76, KSS = 0.53) and DTE imaging (GCNR = 0.81, KSS = 0.62). Further development of the methodology is necessary to distinguish free from adherent MBs. These results demonstrate that neural networks can be trained to detect targeted MBs with DTE-like quality using nondestructive dual-mode data, and can be used to facilitate the safe and real-time translation of UMI to clinical applications.


Assuntos
Aprendizado Profundo , Microbolhas , Animais , Meios de Contraste , Humanos , Camundongos , Imagem Molecular , Ultrassonografia
11.
Invest Radiol ; 55(11): 711-721, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32569010

RESUMO

Early detection of pancreatic ductal adenocarcinoma (PDAC) represents the most significant step toward the treatment of this aggressive lethal disease. Previously, we engineered a preclinical Thy1-targeted microbubble (MBThy1) contrast agent that specifically recognizes Thy1 antigen overexpressed in the vasculature of murine PDAC tissues by ultrasound (US) imaging. In this study, we adopted a single-chain variable fragment (scFv) site-specific bioconjugation approach to construct clinically translatable MBThy1-scFv and test for its efficacy in vivo in murine PDAC imaging, and functionally evaluated the binding specificity of scFv ligand to human Thy1 in patient PDAC tissues ex vivo. MATERIALS AND METHODS: We recombinantly expressed the Thy1-scFv with a carboxy-terminus cysteine residue to facilitate its thioether conjugation to the PEGylated MBs presenting with maleimide functional groups. After the scFv-MB conjugations, we tested binding activity of the MBThy1-scFv to MS1 cells overexpressing human Thy1 (MS1Thy1) under liquid shear stress conditions in vitro using a flow chamber setup at 0.6 mL/min flow rate, corresponding to a wall shear stress rate of 100 seconds, similar to that in tumor capillaries. For in vivo Thy1 US molecular imaging, MBThy1-scFv was tested in the transgenic mouse model (C57BL/6J - Pdx1-Cre; KRas; Ink4a/Arf) of PDAC and in control mice (C57BL/6J) with L-arginine-induced pancreatitis or normal pancreas. To facilitate its clinical feasibility, we further produced Thy1-scFv without the bacterial fusion tags and confirmed its recognition of human Thy1 in cell lines by flow cytometry and in patient PDAC frozen tissue sections of different clinical grades by immunofluorescence staining. RESULTS: Under shear stress flow conditions in vitro, MBThy1-scFv bound to MS1Thy1 cells at significantly higher numbers (3.0 ± 0.8 MB/cell; P < 0.01) compared with MBNontargeted (0.5 ± 0.5 MB/cell). In vivo, MBThy1-scFv (5.3 ± 1.9 arbitrary units [a.u.]) but not the MBNontargeted (1.2 ± 1.0 a.u.) produced high US molecular imaging signal (4.4-fold vs MBNontargeted; n = 8; P < 0.01) in the transgenic mice with spontaneous PDAC tumors (2-6 mm). Imaging signal from mice with L-arginine-induced pancreatitis (n = 8) or normal pancreas (n = 3) were not significantly different between the two MB constructs and were significantly lower than PDAC Thy1 molecular signal. Clinical-grade scFv conjugated to Alexa Fluor 647 dye recognized MS1Thy1 cells but not the parental wild-type cells as evaluated by flow cytometry. More importantly, scFv showed highly specific binding to VEGFR2-positive vasculature and fibroblast-like stromal components surrounding the ducts of human PDAC tissues as evaluated by confocal microscopy. CONCLUSIONS: Our findings summarize the development and validation of a clinically relevant Thy1-targeted US contrast agent for the early detection of human PDAC by US molecular imaging.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Meios de Contraste/metabolismo , Detecção Precoce de Câncer , Neoplasias Pancreáticas/diagnóstico por imagem , Antígenos Thy-1/metabolismo , Ultrassonografia/métodos , Adenocarcinoma/metabolismo , Animais , Linhagem da Célula , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microbolhas , Neoplasias Pancreáticas/metabolismo , Reprodutibilidade dos Testes
12.
Clin Cancer Res ; 26(9): 2140-2150, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31924738

RESUMO

PURPOSE: Human B7-H3 (hB7-H3) is a promising molecular imaging target differentially expressed on the neovasculature of breast cancer and has been validated for preclinical ultrasound (US) imaging with anti-B7-H3-antibody-functionalized microbubbles (MB). However, smaller ligands such as affibodies (ABY) are more suitable for the design of clinical-grade targeted MB. EXPERIMENTAL DESIGN: Binding of ABYB7-H3 was confirmed with soluble and cell-surface B7-H3 by flow cytometry. MB were functionalized with ABYB7-H3 or anti-B7-H3-antibody (AbB7-H3). Control and targeted MB were tested for binding to hB7-H3-expressing cells (MS1hB7-H3) under shear stress conditions. US imaging was performed with MBABY-B7-H3 in an orthotopic mouse model of human MDA-MB-231 coimplanted with MS1hB7-H3 or control MS1WT cells and a transgenic mouse model of breast cancer development. RESULTS: ABYB7-H3 specifically binds to MS1hB7-H3 and murine-B7-H3-expressing monocytes. MBABY-B7-H3 (8.5 ± 1.4 MB/cell) and MBAb-B7-H3 (9.8 ± 1.3 MB/cell) showed significantly higher (P < 0.0001) binding to the MS1hB7-H3 cells compared with control MBNon-targeted (0.5 ± 0.1 MB/cell) under shear stress conditions. In vivo, MBABY-B7-H3 produced significantly higher (P < 0.04) imaging signal in orthotopic tumors coengrafted with MS1hB7-H3 (8.4 ± 3.3 a.u.) compared with tumors with MS1WT cells (1.4 ± 1.0 a.u.). In the transgenic mouse tumors, MBABY-B7-H3 (9.6 ± 2.0 a.u.) produced higher (P < 0.0002) imaging signal compared with MBNon-targeted (1.3 ± 0.3 a.u.), whereas MBABY-B7-H3 signal in normal mammary glands and tumors with B7-H3 blocking significantly reduced (P < 0.02) imaging signal. CONCLUSIONS: MBABY-B7-H3 enhances B7-H3 molecular signal in breast tumors, improving cancer detection, while offering the advantages of a small size ligand and easier production for clinical imaging.


Assuntos
Antígenos B7/metabolismo , Neoplasias da Mama/irrigação sanguínea , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Antígenos B7/imunologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Meios de Contraste/química , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Microbolhas , Imagem Molecular/métodos , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ultrassonografia/métodos
13.
Front Cell Dev Biol ; 7: 159, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31448276

RESUMO

The YAP1/Hippo and p53 pathways are critical protectors of genome integrity in response to DNA damage. Together, these pathways secure cellular adaptation and maintain overall tissue integrity through transcriptional re-programing downstream of various environmental and biological cues generated during normal tissue growth, cell proliferation, and apoptosis. Genetic perturbations in YAP1/Hippo and p53 pathways are known to contribute to the cells' ability to turn rogue and initiate tumorigenesis. The Hippo and p53 pathways cooperate on many levels and are closely coordinated through multiple molecular components of their signaling pathways. Several functional and physical interactions have been reported to occur between YAP1/Hippo pathway components and the three p53 family members, p53, p63, and p73. Primarily, functional status of p53 family proteins dictates the subcellular localization, protein stability and transcriptional activity of the core component of the Hippo pathway, Yes-associated protein 1 (YAP1). In this review, we dissect the critical points of crosstalk between the YAP1/Hippo pathway components, with a focus on YAP1, and the p53 tumor suppressor protein family. For each p53 family member, we discuss the biological implications of their interaction with Hippo pathway components in determining cell fate under the conditions of tissue homeostasis and cancer pathogenesis.

14.
Sci Rep ; 6: 33353, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27634301

RESUMO

Activation of the IRE1α-XBP1 branch of the unfolded protein response (UPR) has been implicated in multiple types of human cancers, including multiple myeloma (MM). Through an in silico drug discovery approach based on protein-compound virtual docking, we identified the anthracycline antibiotic doxorubicin as an in vitro and in vivo inhibitor of XBP1 activation, a previously unknown activity for this widely utilized cancer chemotherapeutic drug. Through a series of mechanistic and phenotypic studies, we showed that this novel activity of doxorubicin was not due to inhibition of topoisomerase II (Topo II). Consistent with its inhibitory activity on the IRE1α-XBP1 branch of the UPR, doxorubicin displayed more potent cytotoxicity against MM cell lines than other cancer cell lines that have lower basal IRE1α-XBP1 activity. In addition, doxorubicin significantly inhibited XBP1 activation in CD138(+) tumor cells isolated from MM patients. Our findings suggest that the UPR-modulating activity of doxorubicin may be utilized clinically to target IRE1α-XBP1-dependent tumors such as MM.


Assuntos
Doxorrubicina/farmacologia , Endorribonucleases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Proteína 1 de Ligação a X-Box/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/química , Etoposídeo/química , Etoposídeo/farmacologia , Humanos , Splicing de RNA/genética , Inibidores da Topoisomerase/farmacologia
15.
Exp Hematol ; 41(6): 547-557.e2, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23435312

RESUMO

Multiple myeloma (MM) cells typically grow in focal lesions, stimulating osteoclasts that destroy bone and support MM. Osteoclasts and MM cells are hypermetabolic. The coenzyme nicotinamide adenine dinucleotide (NAD(+)) is not only essential for cellular metabolism; it also affects activity of NAD-dependent enzymes, such as PARP-1 and SIRT-1. Nicotinamide phosphoribosyltransferase (NAMPT/PBEF/visfatin, encoded by PBEF1) is a rate-limiting enzyme in NAD(+) biosynthesis from nicotinamide. Coculture of primary MM cells with osteoclasts induced PBEF1 upregulation in both cell types. PBEF1 expression was higher in experimental myelomatous bones than in nonmyelomatous bone and higher in MM patients' plasma cells than in healthy donors' counterparts. APO866 is a specific PBEF1 inhibitor known to deplete cellular NAD(+). APO866 at low nanomolar concentrations inhibited growth of primary MM cells or MM cell lines cultured alone or cocultured with osteoclasts and induced apoptosis in these cells. PBEF1 activity and NAD(+) content were reduced in MM cells by APO866, resulting in lower activity of PARP-1 and SIRT-1. The inhibitory effect of APO866 on MM cell growth was abrogated by supplementation of extracellular NAD(+) or NAM. APO866 inhibited NF-κB activity in osteoclast precursors and suppressed osteoclast formation and activity. PBEF1 knockdown similarly inhibited MM cell growth and osteoclast formation. In the SCID-rab model, APO866 inhibited growth of primary MM and H929 cells and prevented bone disease. These findings indicate that MM cells and osteoclasts are highly sensitive to NAD(+) depletion and that PBEF1 inhibition represents a novel approach to target cellular metabolism and inhibit PARP-1 and bone disease in MM.


Assuntos
Citocinas/fisiologia , Mieloma Múltiplo/enzimologia , Proteínas de Neoplasias/fisiologia , Nicotinamida Fosforribosiltransferase/fisiologia , Osteoclastos/enzimologia , Osteólise/enzimologia , Acrilamidas/farmacologia , Animais , Osso e Ossos/patologia , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cocultura , Citocinas/antagonistas & inibidores , Indução Enzimática , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos SCID , Mieloma Múltiplo/complicações , Mieloma Múltiplo/patologia , NAD/metabolismo , NF-kappa B/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Niacinamida/metabolismo , Mononucleotídeo de Nicotinamida/análogos & derivados , Mononucleotídeo de Nicotinamida/farmacologia , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Osteoclastos/fisiologia , Osteólise/etiologia , Osteólise/patologia , Piperidinas/farmacologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Coelhos , Sirtuína 1/metabolismo , Células Tumorais Cultivadas/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA