Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(1): 50-63, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33115805

ABSTRACT

Metabolic dysregulation is a known hallmark of cancer progression, yet the oncogenic signals that promote metabolic adaptations to drive metastatic cancer remain unclear. Here, we show that transcriptional repression of mitochondrial deacetylase sirtuin 3 (SIRT3) by androgen receptor (AR) and its coregulator steroid receptor coactivator-2 (SRC-2) enhances mitochondrial aconitase (ACO2) activity to favor aggressive prostate cancer. ACO2 promoted mitochondrial citrate synthesis to facilitate de novo lipogenesis, and genetic ablation of ACO2 reduced total lipid content and severely repressed in vivo prostate cancer progression. A single acetylation mark lysine258 on ACO2 functioned as a regulatory motif, and the acetylation-deficient Lys258Arg mutant was enzymatically inactive and failed to rescue growth of ACO2-deficient cells. Acetylation of ACO2 was reversibly regulated by SIRT3, which was predominantly repressed in many tumors including prostate cancer. Mechanistically, SRC-2-bound AR formed a repressive complex by recruiting histone deacetylase 2 to the SIRT3 promoter, and depletion of SRC-2 enhanced SIRT3 expression and simultaneously reduced acetylated ACO2. In human prostate tumors, ACO2 activity was significantly elevated, and increased expression of SRC-2 with concomitant reduction of SIRT3 was found to be a genetic hallmark enriched in prostate cancer metastatic lesions. In a mouse model of spontaneous bone metastasis, suppression of SRC-2 reactivated SIRT3 expression and was sufficient to abolish prostate cancer colonization in the bone microenvironment, implying this nuclear-mitochondrial regulatory axis is a determining factor for metastatic competence. SIGNIFICANCE: This study highlights the importance of mitochondrial aconitase activity in the development of advanced metastatic prostate cancer and suggests that blocking SRC-2 to enhance SIRT3 expression may be therapeutically valuable. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/1/50/F1.large.jpg.


Subject(s)
Aconitate Hydratase/metabolism , Biomarkers, Tumor/metabolism , Bone Neoplasms/secondary , Gene Expression Regulation, Neoplastic , Mitochondria/enzymology , Prostatic Neoplasms/pathology , Sirtuin 3/metabolism , Aconitate Hydratase/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Proliferation , Humans , Male , Mice , Mice, Nude , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Coactivator 3/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Sirtuin 3/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Eur J Med Chem ; 144: 493-503, 2018 Jan 20.
Article in English | MEDLINE | ID: mdl-29288946

ABSTRACT

Molecular hybridization is considered as an effective tactic to develop drugs for the treatment of cancer. A series of novel hybrid compounds of isatin and Michael acceptor were designed and synthesized on the basis of association principle. These hybrid compounds were tested for cytotoxic potential against human cancer cell lines namely, BGC-823, SGC-7901 and NCI-H460 by MTT assay. Most compounds showed good anti-growth activities in all tested human cancer cells. SAR and QSAR analysis may provide vital information for the future development of novel anti-cancer inhibitors. Notably, compound 6a showed potent growth inhibition on BGC-823, SGC-7901 and NCI-H460 with the IC50 values of 3.6 ±â€¯0.6, 5.7 ±â€¯1.2, 3.2 ±â€¯0.7 µM, respectively. Besides, colony formation assays, wound healing assays and flow cytometry analysis indicated 6a exhibited a potent anti-growth and anti-migration ability in a concentration-dependence manner through arrested cells in the G2/M phase of cell cycle. Moreover, 6a significantly repressed tumor growth in a NCI-H460 xenograft mouse model. Overall, our findings suggested isatin analogues inspired Michael acceptor may provide promising lead compounds for the development of cancer chemotherapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Isatin/pharmacology , Quantitative Structure-Activity Relationship , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Isatin/chemical synthesis , Isatin/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Tumor Cells, Cultured
3.
J Vis Exp ; (115)2016 09 26.
Article in English | MEDLINE | ID: mdl-27768029

ABSTRACT

Intra-iliac artery (IIA) injection is an efficient approach to introduce metastatic lesions of various cancer cells in animals. Compared to the widely used intra-cardiac and intra-tibial injections, IIA injection brings several advantages. First, it can deliver a large quantity of cancer cells specifically to hind limb bones, thereby providing spatiotemporally synchronized early-stage colonization events and allowing robust quantification and swift detection of disseminated tumor cells. Second, it injects cancer cells into the circulation without damaging the local tissues, thereby avoiding inflammatory and wound-healing processes that confound the bone colonization process. Third, IIA injection causes very little metastatic growth in non-bone organs, thereby preventing animals from succumbing to other vital metastases, and allowing continuous monitoring of indolent bone lesions. These advantages are especially useful for the inspection of progression from single cancer cells to multi-cell micrometastases, which has largely been elusive in the past. When combined with cutting-edge approaches of biological imaging and bone histology, IIA injection can be applied to various research purposes related to bone metastases.


Subject(s)
Bone Neoplasms , Iliac Artery , Neoplasm Metastasis , Animals , Bone and Bones , Disease Models, Animal , Disease Progression , Hindlimb , Humans
4.
Mol Cell Oncol ; 3(3): e1026526, 2016 May.
Article in English | MEDLINE | ID: mdl-27314064

ABSTRACT

We recently discovered that bone micrometastases of breast cancer predominantly reside in the microenvironment termed the "osteogenic niche". The heterotypic adherens junctions between cancer cells and osteogenic cells promote early-stage bone colonization by activating the mTOR pathway in cancer cells. Here, we discuss a few questions raised by these findings.

5.
J Mammary Gland Biol Neoplasia ; 20(3-4): 103-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26490041

ABSTRACT

In breast cancer, the most frequent site of metastasis is bone. Disseminated tumor cells (DTCs) can be detected in the bone marrow of patients by their expression of epithelial oroncogenic markers [1], and the presence and frequency of these DTCs are associated with poor prognosis. However, many of the details behind this process remain elusive, including the biological properties and fates of these apparently indolent cancer cells. To provide pre-clinical models of DTCs, we have developed a procedure that allows for controlled and enhanced delivery of tumor cells to the bone in animal experiments via injection into the iliac artery of the hind limb [2]. To our surprise, we found that most cancer cells became integrated into the solid bone matrix shortly after arriving in the bone, and only a minority can be flushed out with the bone marrow.Here we describe a method that helps to retrieve DTCs homing to the bone in which we achieve an improved recovery of those tumor cells closely associated with the bone microenvironment. In our view it is especially important to analyze these tumor cell subpopulations, as they may take full advantage of growth-, survival- and immune-protective signals provided by neighbor cells. We also show a pilot study on how this approach may be applied to the analysis of cancer dormancy. Our study suggests that the detection and retrieval of DTCs in clinical studies are incomplete because they are conducted exclusively with bone marrow aspirates.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Separation/methods , Specimen Handling/methods , Animals , Bone Marrow/pathology , Cell Survival , Disease Models, Animal , Female , Femur/pathology , Humans , Iliac Artery , Injections, Intra-Arterial , MCF-7 Cells , Matrix Metalloproteinase 8 , Mice , Mice, Inbred BALB C , Mice, Nude , Tibia/pathology
6.
Cancer Cell ; 27(2): 193-210, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25600338

ABSTRACT

Breast cancer bone micrometastases can remain asymptomatic for years before progressing into overt lesions. The biology of this process, including the microenvironment niche and supporting pathways, is unclear. We find that bone micrometastases predominantly reside in a niche that exhibits features of osteogenesis. Niche interactions are mediated by heterotypic adherens junctions (hAJs) involving cancer-derived E-cadherin and osteogenic N-cadherin, the disruption of which abolishes niche-conferred advantages. We elucidate that hAJ activates the mTOR pathway in cancer cells, which drives the progression from single cells to micrometastases. Human data set analyses support the roles of AJ and the mTOR pathway in bone colonization. Our study illuminates the initiation of bone colonization, and provides potential therapeutic targets to block progression toward osteolytic metastases.


Subject(s)
Bone Neoplasms/genetics , Breast Neoplasms/genetics , Osteogenesis/genetics , Tumor Microenvironment/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cadherins/genetics , Cell Line, Tumor , Female , Humans , Neoplasm Staging , TOR Serine-Threonine Kinases/genetics
7.
ASN Neuro ; 2(4): e00040, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20711301

ABSTRACT

Increasing the expression of Hsp70 (heat-shock protein 70) can inhibit sensory neuron degeneration after axotomy. Since the onset of DPN (diabetic peripheral neuropathy) is associated with the gradual decline of sensory neuron function, we evaluated whether increasing Hsp70 was sufficient to improve several indices of neuronal function. Hsp90 is the master regulator of the heat-shock response and its inhibition can up-regulate Hsp70. KU-32 (N-{7-[(2R,3R,4S,5R)-3,4-dihydroxy-5-methoxy-6,6-dimethyl-tetrahydro-2H-pyran-2-yloxy]-8-methyl-2-oxo-2H-chromen-3-yl}acetamide) was developed as a novel, novobiocin-based, C-terminal inhibitor of Hsp90 whose ability to increase Hsp70 expression is linked to the presence of an acetamide substitution of the prenylated benzamide moiety of novobiocin. KU-32 protected against glucose-induced death of embryonic DRG (dorsal root ganglia) neurons cultured for 3 days in vitro. Similarly, KU-32 significantly decreased neuregulin 1-induced degeneration of myelinated Schwann cell DRG neuron co-cultures prepared from WT (wild-type) mice. This protection was lost if the co-cultures were prepared from Hsp70.1 and Hsp70.3 KO (knockout) mice. KU-32 is readily bioavailable and was administered once a week for 6 weeks at a dose of 20 mg/kg to WT and Hsp70 KO mice that had been rendered diabetic with streptozotocin for 12 weeks. After 12 weeks of diabetes, both WT and Hsp70 KO mice developed deficits in NCV (nerve conduction velocity) and a sensory hypoalgesia. Although KU-32 did not improve glucose levels, HbA1c (glycated haemoglobin) or insulin levels, it reversed the NCV and sensory deficits in WT but not Hsp70 KO mice. These studies provide the first evidence that targeting molecular chaperones reverses the sensory hypoalgesia associated with DPN.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Pain Measurement , Sensory Receptor Cells/metabolism , Animals , Axotomy/adverse effects , Axotomy/rehabilitation , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetic Neuropathies/pathology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HSP90 Heat-Shock Proteins/biosynthesis , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Novobiocin/pharmacology , Novobiocin/therapeutic use , Pain Measurement/drug effects , Rats , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology
8.
J Proteome Res ; 9(1): 458-71, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19905032

ABSTRACT

Hyperglycemia-induced mitochondrial dysfunction contributes to sensory neuron pathology in diabetic neuropathy. Although Schwann cells (SCs) also undergo substantial degeneration in diabetic neuropathy, the effect of hyperglycemia on the SC mitochondrial proteome and mitochondrial function has not been examined. Stable isotope labeling with amino acids in cell culture (SILAC) was used to quantify the temporal effect of hyperglycemia on the mitochondrial proteome of primary SCs isolated from neonatal rats. Of 317 mitochondrial proteins identified, about 78% were quantified and detected at multiple time points. Pathway analysis indicated that proteins associated with mitochondrial dysfunction, oxidative phosphorylation, the TCA cycle, and detoxification were significantly increased in expression and over-represented. Assessing mitochondrial respiration in intact SCs indicated that hyperglycemia increased the overall rate of oxygen consumption but decreased the efficiency of coupled respiration. Although a glucose-dependent increase in superoxide production occurs in embryonic sensory neurons, hyperglycemia did not induce a substantial change in superoxide levels in SCs. This correlated with a 1.9-fold increase in Mn superoxide dismutase expression, which was confirmed by immunoblot and enzymatic activity assays. These data support that hyperglycemia alters mitochondrial respiration and can cause remodeling of the SC mitochondrial proteome independent of significant contributions from glucose-induced superoxide production.


Subject(s)
Hyperglycemia/metabolism , Mitochondrial Proteins/metabolism , Proteome/metabolism , Schwann Cells/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism , Analysis of Variance , Animals , Cell Fractionation , Cell Nucleus/metabolism , Isotope Labeling , Mitochondria/metabolism , Oxygen , Rats , Signal Transduction
9.
Cancer Biol Ther ; 7(8): 1226-31, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18487949

ABSTRACT

The second domain of Pseudomonas exotoxin A (PEAII, residues 253-364) has been shown to facilitate translocation of extracellular and vesicular contents into the cytoplasm, and can transport heterologous molecules into target cells. Because full length PEAII may elicit a host immune response, we tried to identify the minimal PEAII translocation motif and use this fragment in combination with an antibody and constitutively active granzyme B (ImmunoGrB) to kill HER2-positive tumor cells. We constructed four ImmunoGrB fusion proteins containing different PEAII deletions and tested their abilities to kill HER2-positive cells. Our data showed that while a complete deletion of PEAII in ImmunoGrB resulted in an inability to kill cancer cells, ImmunoGrBs containing either PEAII (253-358aa) or PEAII (275-358aa) could efficiently kill HER2-positive SK-BR-3 cells. Most interestingly, the construct which contains only a furin cleavage site, named PEAII (275-280aa), could still induce SK-BR-3 apoptosis, although less efficiently. Moreover, delivery of the recombinant proteins by intramuscular plasmid injection led to an apparent tumor regression and prolonged animal survival in a nude mouse xenograft SK-BR-3 tumor model, indicating in vivo antitumor activity of the different PEAII containing ImmunoGrBs. Our results may help in understanding PEAII translocation and may lead to the development of useful tools or alternative therapy.


Subject(s)
Breast Neoplasms/pathology , Genes, erbB-2 , Receptor, ErbB-2/genetics , ADP Ribose Transferases , Bacterial Toxins , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/ultrastructure , Cell Line, Tumor , Cell Survival , Exotoxins , HeLa Cells , Humans , Plasmids , Protein Structure, Tertiary/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Recombinant Proteins/metabolism , Transfection , Virulence Factors , Pseudomonas aeruginosa Exotoxin A
10.
Glia ; 56(8): 877-87, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18338795

ABSTRACT

Neuregulins (NRGs) are growth factors which bind to Erb receptor tyrosine kinases that localize to Schwann cells (SCs). Although NRGs can promote cell survival, mitogenesis, and myelination in undifferentiated SCs, they also induce demyelination of myelinated co-cultures of SCs and dorsal root ganglion (DRG) neurons. We have shown previously that Erb B2 activity increased in premyelinating SCs in response to hyperglycemia, and that this correlated with the downregulation of the protein caveolin-1 (Cav-1). As myelinated SCs undergo substantial degeneration in diabetic neuropathy, we used myelinated SC/DRG neuron co-cultures to determine if hyperglycemia and changes in Cav-1 expression could enhance NRG-induced demyelination. In basal glucose, NRG1 caused a 2.4-fold increase in the number of damaged myelin segments. This damage reached 3.8-fold under hyperglycemic conditions, and was also associated with a robust decrease in the expression of Cav-1 and compact myelin proteins. The loss of Cav-1 and compact myelin proteins following hyperglycemia and NRG treatment was not due to neuronal loss, since the axons remained intact and there was no loss of PGP 9.5, an axonal marker protein. To examine if changes in Cav-1 were sufficient to alter the extent of NRG-induced demyelination, SC/DRG neurons co-cultures were infected with antisense or dominant-negative Cav-1(P132L) adenoviruses. Either antisense-mediated downregulation or mis-localization of endogenous Cav-1 by Cav-1(P132L) resulted in a 1.5- to 2.4-fold increase in NRG-induced degeneration compared to that present in control cultures. These data support that hyperglycemia and changes in Cav-1 are sufficient to sensitize myelinated SC/DRG co-cultures to NRG-induced demyelination.


Subject(s)
Caveolin 1/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/physiopathology , Down-Regulation/physiology , Hyperglycemia/physiopathology , Neuregulin-1 , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Demyelinating Diseases/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Ganglia, Spinal/cytology , Glucose/administration & dosage , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Neuregulin-1/pharmacology , Neurons/drug effects , Neurons/physiology , Rats , Schwann Cells/drug effects , Schwann Cells/physiology , Time Factors , Transfection
11.
J Lipid Res ; 46(8): 1678-91, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15863835

ABSTRACT

Addition of exogenous ceramide causes a significant displacement of cholesterol in lipid raft model membranes. However, whether ceramide-induced cholesterol displacement is sufficient to alter the protein composition of caveolin-enriched lipid raft membranes is unknown. Therefore, we examined whether increasing endogenous ceramide levels with bacterial sphingomyelinase (bSMase) depleted cholesterol and changed the protein composition of caveolin-enriched membranes (CEMs) isolated from immortalized Schwann cells. bSMase increased ceramide levels severalfold and decreased the cholesterol content of detergent-insoluble CEMs by 25-50% within 2 h. To examine the effect of ceramide on the protein composition of the CEMs, we performed a quantitative proteomic analysis using stable isotope labeling of cells in culture and matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Although ceramide rapidly depleted lipid raft cholesterol, the levels of the cholesterol binding protein caveolin-1 (Cav-1) decreased by 25% only after 8 h. Importantly, replenishing the cells with cholesterol rapidly reversed the loss of Cav-1 from the CEMs. Ceramide-induced cholesterol depletion increased the association of 5'-nucleotidase and ATP synthase beta-subunit with the CEMs but had a minimal effect on changing the abundance of other lipid raft proteins, such as flotillin-1 and G-proteins. These results suggest that the ceramide-induced loss of cholesterol from CEMs may contribute to altering the lipid raft proteome.


Subject(s)
Ceramides/metabolism , Cholesterol/metabolism , Membrane Microdomains/chemistry , 5'-Nucleotidase/analysis , Animals , Animals, Newborn , Membrane Proteins/analysis , Mitochondrial Proton-Translocating ATPases/analysis , Proteomics , Rats , Rats, Sprague-Dawley
12.
Ai Zheng ; 24(2): 160-5, 2005 Feb.
Article in Chinese | MEDLINE | ID: mdl-15694025

ABSTRACT

BACKGROUND & OBJECTIVE: Apoptosis is a kind of evolutional high conservative cell death. Transferring high active pro-apoptotic molecules into cancer cells to induce apoptosis is a potential strategy for cancer gene therapy. Based on our previous generation of reconstructed human caspase-8, which can continuously induce apoptosis of cervical cancer cell line HeLa, by reversing its large and small subunits, this study was designed to investigate the pro-apoptotic efficiencies of 3 reconstructed human caspase-8 (Casp8CD, Rev8, and Rev8L) on HeLa cells, and to explore the feasibility of reconstructed human caspase-8 as potential apoptosis-inducing candidates. METHODS: The eukaryotic expression vectors pIRES2-EGFP carrying Casp8CD, Rev8, and Rev8L genes were transfected into HeLa cells, and breast cancer MCF-7 cells. Expressions and pro-apoptotic effects of Casp8CD, Rev8, and Rev8L genes were observed under fluorescent microscope, and their pro-apoptotic efficiencies were assessed by MTT assay and cells counting. The flexibilities of linking-peptides between subunits of Rev8 and Rev8L were analyzed by bioinformatics. RESULTS: Expressions of the 3 reconstructed caspase-8 genes were observed under fluorescent microscope, and the HeLa and MCF-7 cells expressing Rev8 or Rev8L genes displayed typical apoptotic volume decrease (AVD). MTT assay showed that compared with control cells, A(570) values of Rev8- and Rev8L-transfected cells began to decrease 20 h after transfection. Cell counting results indicated that cell death ratio of Casp8CD-, Rev8-, and Rev8L-transfected cells were 16.9%, 52.3%, and 47.7%, respectively, 24 h after transfection; and 12.9%, 51.6%,and 61.2%,respectively,48 h after transfection. Bioinformatics analysis showed that the linking-peptides between subunits of Rev8 and Rev8L were flexible. CONCLUSION: Rev8 and Rev8L molecules have similar pro-apoptotic effects and efficiencies, but over-expressed Casp8CD had no significant pro-apoptotic effects.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Caspases/physiology , Breast Neoplasms/metabolism , Caspase 8 , Caspases/biosynthesis , Caspases/genetics , Cell Line, Tumor , Female , Genetic Vectors , HeLa Cells , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transfection
13.
J Pharmacol Exp Ther ; 313(2): 485-91, 2005 May.
Article in English | MEDLINE | ID: mdl-15608075

ABSTRACT

Diabetic peripheral neuropathy (DPN) is a frequent and potentially traumatic complication in diabetic individuals. The chronic nature of diabetes and its associated hyperglycemic episodes initiate a complex and inter-related series of metabolic and vascular insults that contribute to the polygenic etiology of DPN. One contributing factor in DPN is an altered neurotrophism that results from changes in the synthesis and expression of neurotrophins, insulin-like growth factor, and various cytokine-like growth factors that can directly act upon distinct subpopulations of sensory and motor neurons. Although considerable effort has focused upon examining growth factor signaling in hyperglycemically stressed neurons, myelin-forming Schwann cells also undergo substantial degenerative changes in DPN. However, scant attention has been devoted to understanding the effect of hyperglycemia on the response of Schwann cells to growth factors critical to their function. Neuregulins are gliotrophic growth factors that signal through members of the Erb B receptor-tyrosine kinase family. The neuregulin/Erb B ligand-receptor cassette can differentially influence the response of Schwann cells throughout their development by regulating cell survival, mitogenesis, and differentiation. The activity of Erb B receptors may also be affected by their interaction with caveolin-1, a protein marker of caveolae ("little caves"). However, whether neuregulin signaling may be directly or indirectly altered under conditions of hyperglycemic stress and contribute to the physiological progression of DPN is unknown. This brief review will provide a perspective on a putative role of changes in the caveolar proteome of Schwann cells in contributing to an "altered neuregulinism" in DPN.


Subject(s)
Caveolae/metabolism , Caveolae/pathology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Nerve Growth Factors/physiology , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Animals , Diabetic Neuropathies/physiopathology , Humans , Peripheral Nerves/physiopathology
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 20(5): 621-4, 2004 Sep.
Article in Chinese | MEDLINE | ID: mdl-15367364

ABSTRACT

AIM: To construct inducible expression vector for human granzyme B gene and express it in Hela cell line. METHODS: Human active granzyme B gene was obtained by PCR and cloned into the inducible expression vector pIND. The resulting expression vector, together with a helper plasmid pVgRXR, was stably transfected into Hela cells using Lipofectamine 2000. The transfected cells were selected in medium containing G418 and zeocin. The resistant cells were induced with ponasterone A, and the optimal concentration of ponasterone A and time of induction were determined by immunocytochemical staining. Then the effects of the expressed granzyme protein on Hela cells were detected by MTT colorimetry and cytoskeletal staining. RESULTS: The Hela cells that inducibly expressed human active granzyme B were obtained. Induction with 30 micromol/L ponasterone A for 5 days caused the strongest expression of granzyme B. The induced cells appeared as either multinucleate giant cells or pyknotic small cells, and their growth was inhibited. Further analysis demonstrated cytoskeletal abnormality of multinucleate giant cells. CONCLUSION: The establishment of inducible expression system for active granzyme B lays the foundation for further research on biological function of granzyme B.


Subject(s)
Ecdysterone/analogs & derivatives , Gene Expression Regulation, Enzymologic/drug effects , Serine Endopeptidases/biosynthesis , Cell Proliferation , Cytoskeleton , Ecdysterone/pharmacology , Genetic Vectors/genetics , Granzymes , HeLa Cells , Humans , Immunohistochemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/physiology , Transfection
15.
J Immunol ; 173(1): 61-7, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15210759

ABSTRACT

Clinical studies have suggested that human epidermal growth factor receptor-2 (HER2) provide a useful target for antitumor therapy. We previously described the generation of a chimeric HER2-targeted immunocasp-3 protein. In this study, we extend the repertoire of chimeric proapoptotic proteins with immunocasp-6, a construct that comprises a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A, and an active caspase-6, which can directly cleave lamin A leading to nucleus damage and inducing programmed cell death. We demonstrate that the secreted immunocasp-6 molecule selectively recognizes and induces apoptosis in HER2-overexpressing tumor cells in vitro, but not in cells with undetectable HER2. The immunocasp-6 gene was next transferred into BALB/c athymic mice bearing human breast SK-BR-3 tumors by i.m. injection of liposome-encapsulated vectors, by intratumor injection of adenoviral vectors, or by i.v. injection of PBMC modified by retroviral infection. Regardless of the method used, expression of immunocasp-6 suppressed tumor growth and prolonged animal survival significantly. Our data show that the chimeric immunocasp-6 molecule can recognize HER2-positive tumor cells, promptly attack their nucleus, and induce their apoptotic death, suggesting the potential of this strategy for the treatment of human cancers that overexpress HER2.


Subject(s)
Antibodies/therapeutic use , Caspases/genetics , Genetic Therapy , Neoplasms, Experimental/therapy , Receptor, ErbB-2/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , ADP Ribose Transferases/therapeutic use , Adenoviridae/genetics , Animals , Apoptosis , Bacterial Toxins/therapeutic use , Caspase 6 , Cell Line, Tumor , Exotoxins/therapeutic use , Humans , Jurkat Cells , Liposomes , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/chemistry , Receptor, ErbB-2/analysis , Receptor, ErbB-2/immunology , Retroviridae/genetics , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 20(1): 19-22, 2004 Jan.
Article in Chinese | MEDLINE | ID: mdl-15182613

ABSTRACT

AIM: To explore the expression of the truncated bid gene and its pro-apoptotic effect on Hela cells. METHODS: A full-length human bid gene was cloned by RT-PCR and confirmed by sequence analysis. By deleting the 60 amino acids of N-terminal the truncated bid (tbid) gene was obtained and then inserted into the eukaryotic expression vector pIRES2-EGFP. After the tbid gene was transfected into Hela cells under lipofectamine mediation, the effect of target gene expression on morphology and growth of Hela cells were observed under fluorescence and electron microscopes and analysed by TUNEL staining. RESULTS: pIRES2-EGFP containing tbid gene was constructed successfully. After Hela cells were transfected with GFP expression vector of tbid gene, tbid was expressed which was followed by decreased cell fluorescence intensity, poor cell growth, and cell death. Cell shrinkage and nuclear condensation, typical apoptotic characteristics, were observed by electron microscope observation and Tunel staining. CONCLUSION: The expression of tbid can effectively induce apoptosis of Hela cells.


Subject(s)
Apoptosis , Carrier Proteins/genetics , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/physiology , Cell Division , Cloning, Molecular , Green Fluorescent Proteins , HeLa Cells , Humans , In Situ Nick-End Labeling , Luminescent Proteins/genetics , Polymerase Chain Reaction , Transfection
17.
J Biol Chem ; 279(20): 21343-8, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15004021

ABSTRACT

Targeted cell killing is required for effective treatment of cancers. We previously described the generation of a chimeric immunocasp-3 protein and its potent selective antitumor activity (Jia, L. T., Zhang, L. H., Yu, C. J., Zhao, J., Xu, Y. M., Gui, J. H., Jin, M., Ji, Z. L., Wen, W. H., Wang, C. J., Chen, S. Y., and Yang, A. G. (2003) Cancer Res. 63, 3257-3262). Here we extend the repertoire of another chimeric pro-apoptotic protein immunoGrB, which comprises an anti-HER2 single-chain antibody, a Pseudomonas exotoxin A translocation domain and active granzyme B. Human lymphoma Jurkat cells transfected with the immunoGrB gene expression vector were able to produce and secrete the chimeric protein. The immunoGrB molecule selectively recognized and destroyed HER2-overexpressing tumor cells both in vitro and in nude mouse after intramuscular injection of the immunoGrB expression plasmid. Further in vivo study showed that intravenous administration of immunoGrB gene-modified lymphocytes led to suppression of HER2-overexpressing tumor growth and prolonged animal survival because of continuous secretion of immunoGrB molecules into blood and lymph fluid. These results demonstrate that the chimeric immunoGrB molecule, which is capable of antibody-directed targeting and granzyme B-mediated killing, has therapeutic potential against HER2 tumors, especially in cases in which caspase-dependent apoptosis is inhibited.


Subject(s)
Antibodies/toxicity , Breast Neoplasms/genetics , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/toxicity , Serine Endopeptidases/toxicity , Animals , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Female , Granzymes , Humans , Mice , Mice, Nude , Transfection , Transplantation, Heterologous
18.
Sheng Wu Gong Cheng Xue Bao ; 20(4): 501-6, 2004 Jul.
Article in Chinese | MEDLINE | ID: mdl-15968978

ABSTRACT

Translocating protein and translocating peptides have therapeutic potential against tumors by exposing the cytotoxic domains of toxic proteins to the cell cytosol. The aim of this study is to investigate the effect of N-terminally fused PE translocating peptides on granzyme B (GrBa) activity. PE II-GrBa fusion protein genes were constructed by replacing N-terminal signal and acidic dipeptide sequence of human granzyme B gene with two truncated translocating sequences of Pseudomonas exotoxin A (PE II aa 280-364/358) by recombinant PCR, and then cloned into pIND inducible expression vector. The resulting pIND-PE II-GrBa expression vectors were co-transfected with assistant plasmid pVgRXR into HeLa cells through lipofectamine, followed by selection on G418 and zeocin. The resistant cells were collected and induced with ponasterone A. Western blot analysis demonstrated that ponasterone A induction caused the expression of PE II-GrBa fusion proteins, and indirect immunofluorescence detected giant sized multinucleated cells, suggesting cytoskeletal and mitotic abnormalities as reported in our previous studies. Western blot, enzymatic activity assay and cell counting analysis indicated that two types of PE II-GrBa fusion proteins were capable of cleaving both endogenous and exogenous substrates of granzyme B, and inhibiting the growth of cells. The PE II (aa 280-358)-GrBa was shown to have higher serine protease activity and stronger growth inhibitory effect. Such inhibition was presumably associated with G2 arrest as determined by cell cycle analysis. These data prove that PE II-GrBa fusion proteins have cell inhibitory effect similar to GrBa, and that the shorter PE-derived peptide exerts less influence on GrBa activity. This study helps to optimize the construction of recombinant protein comprising translocating peptides and cytotoxic molecules for tumor cell killing.


Subject(s)
ADP Ribose Transferases/pharmacology , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Granzymes/pharmacology , Recombinant Fusion Proteins/pharmacology , Virulence Factors/pharmacology , ADP Ribose Transferases/genetics , Bacterial Toxins/genetics , Cell Proliferation/drug effects , Exotoxins/genetics , Granzymes/genetics , HeLa Cells , Humans , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
19.
Zhonghua Yi Xue Za Zhi ; 83(7): 564-8, 2003 Apr 10.
Article in Chinese | MEDLINE | ID: mdl-12887745

ABSTRACT

OBJECTIVE: To investigate the targeted killing effect to SKBr3 cells due to the expression of a secreted fusion protein consisting of anti-erbB2 antibody and reversed caspase-3. METHODS: A recombinant plasmid pCMV-e23scFv-PEII-revcasp 3 was constructed by subcloning reversed caspase-3 gene to the downstream of anti-erbB2 antibody and transfected into Jurkat cells. The cell lines which secreted expressing fusion protein stably were selected. The fusion protein in media was detected by ELISA and the media was used to culture human breast cancer SKBr3 cells. The recombinant plasmids with liposomes was administrated to BALB/C nude mouses bearing SKBr3 tumor by intramuscular injection. The targetting effect of the recombinant fusion protein caspase-3 was detected by indirect immunofluorescence staining. RESULTS: Fusion protein can be expressed and secreted by Jurkat cells stably and kill SKBr3 cells. Significant prolonged survival time (prolonged by 72%) and inhibition of tumor growth in vivo (within inhibition ratio of 77%) were seen in the group administered with recombinant plasmids. Indirect immunofluorescence staining showed that the recombinant fusion protein caspase-3 has targetting effect. CONCLUSION: Secreted expression of the fusion protein consisting of anti-erbB2 antibody and reversed caspase-3 can targetedly induce SKBr3 cells to death.


Subject(s)
Antibodies/genetics , Caspases/genetics , Genetic Therapy , Neoplasms, Experimental/therapy , Receptor, ErbB-2/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Animals , Caspase 3 , Humans , Jurkat Cells , Mice , Mice, Inbred BALB C
20.
Cancer Res ; 63(12): 3257-62, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810656

ABSTRACT

In this study, a novel approach to antitumor therapy was devised by generating a chimeric tumor-targeted killer protein, referred to as immunocasp-3, that comprises a single-chain anti-erbB2/HER2 antibody with a NH(2)-terminal signal sequence, a Pseudomonas exotoxin A translocation domain, and a constitutively active caspase-3 molecule. In principle, cells transfected with the immunocasp-3 gene would express and secrete the chimeric protein, which then binds to HER2-overexpressing tumor cells. Subsequent cleavage of the constitutively active capase-3 domain from the immunocasp-3 molecule and its release from internalized vesicles would lead to apoptotic tumor cell death. To test this strategy, we transduced human lymphoma Jurkat cells with a chimeric immunocasp-3 gene expression vector and showed that they not only expressed and secreted the fusion protein but also selectively killed tumor cells overexpressing HER2 in vitro. i.v. injection of the transduced Jurkat cells led to tumor regression in a mouse xenograft model because of continuous secretion of immunocasp-3 by the transduced cells. The growth of HER2-positive tumor cells in this model was inhibited by i.m. as well as intratumor injection of immunocasp-3 expression plasmid DNA, indicating that the immunocasp-3 molecules secreted by transfected cells have systematic antitumor activity. We conclude that the immunocasp-3 molecule, combining the properties of a tumor-specific antibody with the proapoptotic activity of a caspase, has potent and selective antitumor activity, either as cell-based therapy or as a DNA vaccine. These findings provide a compelling rationale for therapeutic protocols designed for erbB2/HER2-positive tumors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antigens, Neoplasm/immunology , Apoptosis/drug effects , Caspases/therapeutic use , Immunoconjugates/therapeutic use , Immunoglobulin Variable Region/therapeutic use , Neoplasm Proteins/immunology , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/therapeutic use , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Caspase 3 , Caspases/pharmacology , Enzyme Activation , Enzyme Induction , Female , HeLa Cells , Humans , Immunoconjugates/pharmacology , Immunoglobulin Variable Region/pharmacology , Jurkat Cells/metabolism , Jurkat Cells/transplantation , Mice , Molecular Sequence Data , Ovarian Neoplasms/pathology , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...