Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Neuroinflammation ; 16(1): 174, 2019 Aug 31.
Article in English | MEDLINE | ID: mdl-31472686

ABSTRACT

BACKGROUND: Oxidative stress, inflammation, and endoplasmic reticulum (ER) stress play a major role in the pathogenesis of neonatal hypoxic-ischemic (HI) injury. ER stress results in the accumulation of unfolded proteins that trigger the NADPH-P450 reductase (NPR) and the microsomal monooxygenase system which is composed of cytochrome P450 members (CYP) generating reactive oxygen species (ROS) as well as the release of inflammatory cytokines. We explored the role of Bax Inhibitor-1 (BI-1) protein, encoded by the Transmembrane Bax inhibitor Motif Containing 6 (TMBIM6) gene, in protection from ER stress after HI brain injury. BI-1 may attenuate ER stress-induced ROS production and release of inflammatory mediators via (1) disruption of the NPR-CYP complex and (2) upregulation of Nrf-2, a redox-sensitive transcription factor, thus promoting an increase in anti-oxidant enzymes to inhibit ROS production. The main objective of our study is to evaluate BI-1's inhibitory effects on ROS production and inflammation by overexpressing BI-1 in 10-day-old rat pups. METHODS: Ten-day-old (P10) unsexed Sprague-Dawley rat pups underwent right common carotid artery ligation, followed by 1.5 h of hypoxia. To overexpress BI-1, rat pups were intracerebroventricularly (icv) injected at 48 h pre-HI with the human adenoviral vector-TMBIM6 (Ad-TMBIM6). BI-1 and Nrf-2 silencing were achieved by icv injection at 48 h pre-HI using siRNA to elucidate the potential mechanism. Percent infarcted area, immunofluorescent staining, DHE staining, western blot, and long-term neurobehavior assessments were performed. RESULTS: Overexpression of BI-1 significantly reduced the percent infarcted area and improved long-term neurobehavioral outcomes. BI-1's mediated protection was observed to be via inhibition of P4502E1, a major contributor to ROS generation and upregulation of pNrf-2 and HO-1, which correlated with a decrease in ROS and inflammatory markers. This effect was reversed when BI-1 or Nrf-2 were inhibited. CONCLUSIONS: Overexpression of BI-1 increased the production of antioxidant enzymes and attenuated inflammation by destabilizing the complex responsible for ROS production. BI-1's multimodal role in inhibiting P4502E1, together with upregulating Nrf-2, makes it a promising therapeutic target.


Subject(s)
Apoptosis Regulatory Proteins/administration & dosage , Cytochrome P-450 Enzyme System/metabolism , Gene Transfer Techniques , Hypoxia-Ischemia, Brain/metabolism , Membrane Proteins/administration & dosage , NADPH-Ferrihemoprotein Reductase/metabolism , NF-E2-Related Factor 2/biosynthesis , Adenoviridae/genetics , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/genetics , Brain/growth & development , Cytochrome P-450 Enzyme System/genetics , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/prevention & control , Male , Membrane Proteins/genetics , NADPH-Ferrihemoprotein Reductase/genetics , NF-E2-Related Factor 2/genetics , Rats , Up-Regulation/physiology
2.
Int J Mol Sci ; 20(1)2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30621194

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is an endogenous neuropeptide also secreted by non-neural cells, including chondrocytes. PACAP signaling is involved in the regulation of chondrogenesis, but little is known about its connection to matrix turnover during cartilage formation and under cellular stress in developing cartilage. We found that the expression and activity of hyaluronidases (Hyals), matrix metalloproteinases (MMP), and aggrecanase were permanent during the course of chondrogenesis in primary chicken micromass cell cultures, although protein levels changed daily, along with moderate and relatively constant enzymatic activity. Next, we investigated whether PACAP influences matrix destructing enzyme activity during oxidative and mechanical stress in chondrogenic cells. Exogenous PACAP lowered Hyals and aggrecanase expression and activity during cellular stress. Expression and activation of the majority of cartilage matrix specific MMPs such as MMP1, MMP7, MMP8, and MMP13, were also decreased by PACAP addition upon oxidative and mechanical stress, while the activity of MMP9 seemed not to be influenced by the neuropeptide. These results suggest that application of PACAP can help to preserve the integrity of the newly synthetized cartilage matrix via signaling mechanisms, which ultimately inhibit the activity of matrix destroying enzymes under cellular stress. It implies the prospect that application of PACAP can ameliorate articular cartilage destruction in joint diseases.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Chondrocytes/drug effects , Oxidative Stress , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Stress, Mechanical , Animals , Apoptosis Regulatory Proteins/administration & dosage , Cartilage/drug effects , Cartilage/metabolism , Cell Culture Techniques , Chick Embryo , Chondrocytes/metabolism , Endopeptidases/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Hyaluronoglucosaminidase/metabolism , Hydrogen Peroxide/pharmacology , Matrix Metalloproteinases/metabolism , Oxidants/pharmacology
3.
Wound Repair Regen ; 26(6): 413-425, 2018 11.
Article in English | MEDLINE | ID: mdl-30155987

ABSTRACT

Radiation therapy is an effective treatment strategy for many types of cancer but is limited by its side effects on normal tissues, particularly the skin, where persistent and progressive fibrotic changes occur and can impair wound healing. In this study, we attempted to mitigate the effects of irradiation on skin using a novel transcutaneous topical delivery system to locally inhibit p53 up-regulated modulator of apoptosis (PUMA) gene expression with small interfering RNA (siRNA). In an isolated skin irradiation model, the dorsal skin of C57 wild-type mice was irradiated. Prior to irradiation, PUMA and nonsense siRNA were applied via a novel hydrogel formulation to dorsal skin and reapplied weekly. Skin was harvested at multiple time points to evaluate dermal siRNA penetration, mRNA expression, protein expression, dermal thickness, subcutaneous fat, stiffness, vascular hypertrophy, SCAR index, and reactive oxygen species (ROS) generation. Murine skin treated with topical PUMA siRNA via optimized hydrogel formulation demonstrated effective PUMA inhibition in irradiated tissue at 3-4 days. Tissue stiffness, dermal thickness, vascular hypertrophy, SCAR index, ROS levels, and mRNA levels of MnSOD and TGF-ß were all significantly reduced with siPUMA treatment compared to nonsense controls. Subcutaneous fat area was significantly increased, and levels of SMAD3 and Phospho-SMAD3 expression were unchanged. These results show that PUMA expression can be effectively silenced in vivo using a novel hydrogel lipoplex topical delivery system. Moreover, cutaneous PUMA inhibition mitigates radiation induced changes in tissue character, restoring a near-normal phenotype independent of SMAD3 signaling.


Subject(s)
Apoptosis Regulatory Proteins/administration & dosage , Apoptosis Regulatory Proteins/pharmacology , Radiation Injuries, Experimental/prevention & control , Signal Transduction/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Wound Healing/drug effects , Administration, Cutaneous , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Cells, Cultured , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/pathology , Gels , Gene Expression Regulation , Mice , Mice, Inbred C57BL
4.
Clin Exp Pharmacol Physiol ; 45(2): 140-145, 2018 02.
Article in English | MEDLINE | ID: mdl-28945941

ABSTRACT

Multiple myeloma (MM) is one of the most common hematological malignancies and characterized by the clonal accumulation of malignant plasma cells. Significant progress has been made in MM treatment recently, while MM still remains incurable. Our previous studies showed that the recombined human programmed cell death 5 (rhPDCD5) can promote MM apoptosis induced by dexamethasone (Dex). Here, we expanded the findings by showing that the rhPDCD5 alone could not induce an obvious growth inhibition of U266 cells (a MM cell line). Of note, with the combination of dexamethasone (Dex), the growth of MM cells was significantly inhibited and accompanied with the cell cycle arrest in G0/G1. For mechanism study, we found that the combination treatment of rhPDCD5 plus Dex downregulated the mRNA and protein expressions of Wnt effectors including ß-catenin, ß-catenin (Ser675), TCF4, survivin and c-Myc when compared to Dex only. Moreover, the activation of WNT pathway induced by LiCl can also be inhibited by this combination treatment. Taken together, our study demonstrated that the combination of rhPDCD5 and Dex can suppress the proliferation of multiple myeloma cells partially via inhibiting the WNT signalling pathway.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Dexamethasone/pharmacology , Multiple Myeloma/drug therapy , Neoplasm Proteins/pharmacology , Wnt Proteins/metabolism , Apoptosis Regulatory Proteins/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Dexamethasone/administration & dosage , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lithium Chloride/pharmacology , Neoplasm Proteins/administration & dosage , Recombinant Proteins , Wnt Proteins/genetics
5.
Tumour Biol ; 39(7): 1010428317716689, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28720068

ABSTRACT

Wilms' tumor is associated with a high treatment success rate, but there is still a risk of recurrence. Cisplatin, which is one of the chemotherapeutic agents used for its treatment, is associated with a very high rate of resistance. Par-4 (prostate apoptosis response 4) is a tumor suppressor, which is capable of sensitizing tumor cells to chemotherapy. Therefore, the aim of this study was to determine whether combined treatment with Par-4 and cisplatin is effective for inhibiting growth of Wilms' tumor. Wilms' tumor and control cell samples were collected and analyzed by immunofluorescence assay and immunohistochemistry. Total proteins extracted from cultured cells were analyzed using western blotting and flow cytometry. In addition, a mouse xenograft model was established. We discovered significantly low expression of Par-4 in the tumor tissue, which was positively correlated with high expression of GRP78 (glucose-regulated protein 78). In addition, we found that ectopic Par-4 co-localized with cell surface GRP78 and induced high expression of the endoplasmic reticulum proteins ATF4 and BAX, which activated the endoplasmic reticulum apoptosis pathway. Moreover, treatment with ectopic Par-4 and cisplatin suppressed xenograft growth in nude mice. In conclusion, our results showed that Par-4 overexpression and cisplatin had a synergistic effect on SK-NEP-1 cells, as a result of which cell growth was inhibited and cellular apoptosis was induced. Thus, in vitro and in vivo upregulation of Par-4 expression is indispensable for the trafficking of GRP78 to the cell membrane and subsequent apoptosis of cancer cells.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Heat-Shock Proteins/genetics , Wilms Tumor/drug therapy , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/genetics , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/biosynthesis , Humans , Mice , Signal Transduction/drug effects , Transfection , Wilms Tumor/genetics , Wilms Tumor/pathology , Xenograft Model Antitumor Assays
6.
Audiol Neurootol ; 20(3): 172-82, 2015.
Article in English | MEDLINE | ID: mdl-25872149

ABSTRACT

Inhibition of cochlear N-methyl-D-aspartate (NMDA) receptors with AM-101, a small molecule antagonist delivered by intratympanic injection, represents a novel approach to treat acute tinnitus triggered by glutamate excitotoxicity. An earlier double-blind, randomized, placebo-controlled phase II clinical trial (TACTT0) had demonstrated a significant and dose-dependent improvement in tinnitus triggered by acute acoustic trauma or otitis media from baseline to day 90. A second phase II trial (TACTT1) now sought to evaluate the most appropriate dose regimen for this treatment. Outcomes from the TACTT1 trial showed no significant difference in tinnitus improvement between a single-dose treatment and a dose regimen comprising three doses over 2 weeks. Taken together, three injections over 3 consecutive days showed the best results in the two phase II trials, suggesting that repeated and concentrated inhibition of cochlear NMDA receptors provides best treatment effects, while keeping the procedural impact on patients short.


Subject(s)
Apoptosis Regulatory Proteins/administration & dosage , Tinnitus/drug therapy , Adolescent , Adult , Apoptosis Regulatory Proteins/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Injection, Intratympanic , Male , Middle Aged , Treatment Outcome , Tympanic Membrane/drug effects , Young Adult
7.
Stroke ; 42(3): 764-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21273567

ABSTRACT

BACKGROUND AND PURPOSE: Osteopontin (OPN) is neuroprotective in ischemic brain injuries in adult experimental models; therefore, we hypothesized that OPN would provide neuroprotection and improve long-term neurological function in the immature brain after hypoxic-ischemic (HI) injury. METHODS: HI was induced by unilateral ligation of the right carotid artery followed by hypoxia (8% O(2) for 2 hours) in postnatal Day 7 rats. OPN (0.03 µg or 0.1 µg) was injected intracerebroventricularly at 1 hour post-HI. Temporal expression of endogenous OPN was evaluated in the normal rat brain at the age of 0, 4, 7, 11, 14, and 21 days and in the ipsilateral hemisphere after HI. The effects of OPN were evaluated using 2-3-5-triphenyl tetrazolium chloride staining, apoptotic cell death assay, and cleaved caspase-3 expression. Neurological function was assessed by the Morris water maze test. RESULTS: Endogenous OPN expression in the brain was the highest at the age of 0 day with continuous reduction until the age of 21 days during development. After HI injury, endogenous OPN expression was increased and peaked at 48 hours. Exogenous OPN decreased infarct volume and improved neurological outcomes 7 weeks after HI injury. OPN-induced neuroprotection was blocked by an integrin antagonist. CONCLUSIONS: OPN-induced neuroprotection was associated with cleaved-caspase-3 inhibition and antiapoptotic cell death. OPN treatment improved long-term neurological function against neonatal HI brain injury.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Disease Models, Animal , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Osteopontin/physiology , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/administration & dosage , Apoptosis Regulatory Proteins/therapeutic use , Caspase 3/metabolism , Caspase 3/physiology , Caspase Inhibitors , Cell Death/drug effects , Cell Death/physiology , Osteopontin/administration & dosage , Osteopontin/therapeutic use , Rats
8.
J Immunol ; 183(9): 5983-90, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812188

ABSTRACT

Sepsis, a highly lethal systemic inflammatory syndrome, is associated with increases of proinflammatory cytokines (e.g., TNF-alpha, HMGB1) and the accumulation of apoptotic cells that have the potential to be detrimental. Depending on the timing and tissue, prevention of apoptosis in sepsis is beneficial; however, thwarting the development of secondary necrosis through the active removal of apoptotic cells by phagocytosis may offer a novel anti-sepsis therapy. Immature dendritic cells (IDCs) release exosomes that contain milk fat globule EGF factor VIII (MFGE8), a protein required to opsonize apoptotic cells for phagocytosis. In an experimental sepsis model using cecal ligation and puncture, we found that MFGE8 levels decreased in the spleen and blood, which was associated with impaired apoptotic cell clearance. Administration of IDC-derived exosomes promoted phagocytosis of apoptotic cells and significantly reduced mortality. Treatment with recombinant MFGE8 was equally protective, whereas MFGE8-deficient mice suffered from increased mortality. IDC exosomes also attenuated the release of proinflammatory cytokines in septic rats. Liberation of HMGB1, a nuclear protein that contributes to inflammation upon release from unengulfed apoptotic cells, was prevented by MFGE8-mediated phagocytosis in vitro. We conclude that IDC-derived exosomes attenuate the acute systemic inflammatory response in sepsis by enhancing apoptotic cell clearance via MFGE8.


Subject(s)
Antigens, Surface/physiology , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Exosomes/immunology , Exosomes/metabolism , Sepsis/metabolism , Sepsis/therapy , Animals , Antigens, Surface/administration & dosage , Apoptosis Regulatory Proteins/administration & dosage , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/physiology , Cells, Cultured , Dendritic Cells/pathology , Inflammation Mediators/administration & dosage , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Milk Proteins/administration & dosage , Milk Proteins/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Sepsis/immunology , Sepsis/pathology
9.
Mol Biol Rep ; 38(4): 2323-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21080083

ABSTRACT

Chemotherapy is the major therapy for cancer in clinic. However, chemotherapeutic agents can harm the other tissues/organs besides cancer. Thus, there are great interests in protecting the innocents by the transfer of protective genes. There are two problems to be solved, one is the selection of protective genes and the other is the orientation of the exotic genes. Recent researches demonstrated that the principal mechanism of chemotherapeutics was through apoptosis. Hereby, introduction of anti-apoptosis genes might interrupt the processes of apoptosis to avoid side effect from chemotherapeutics. On the other hand, tissue-specific promoters, which control gene expression in a tissue-specific manner, might be an alternative tool to guarantee the location of target genes. In this research, we applied gene therapy to chemoprotection using anti-apoptosis gene survivin and ovarian-specific promoter OSP-2. The results showed that OSP-2 could specifically drive the expression of survivin in ovarian cells and survivin could protect cells via inhibiting apoptosis. This might put a light on the future of chemoprotective gene therapy.


Subject(s)
Apoptosis Regulatory Proteins/administration & dosage , Apoptosis/drug effects , Drug-Related Side Effects and Adverse Reactions , Gene Targeting/methods , Genetic Therapy/methods , Neoplasms/drug therapy , Analysis of Variance , Animals , Apoptosis Regulatory Proteins/genetics , CHO Cells , Cricetinae , Cricetulus , Flow Cytometry , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Immunohistochemistry , Inhibitor of Apoptosis Proteins/administration & dosage , Promoter Regions, Genetic/genetics , Survivin , Tetrazolium Salts , Thiazoles , Transfection
10.
PLoS One ; 16(4): e0249838, 2021.
Article in English | MEDLINE | ID: mdl-33891625

ABSTRACT

Reperfusion injury following cold and warm ischemia (IRI) is unavoidable during kidney transplantation and contributes to delayed graft function (DGF) and premature graft loss. Death of tubular epithelial cells (TECs) by necrosis during IRI releases pro-inflammatory mediators (e.g. HMGB1), propagating further inflammation (necroinflammation) and tissue damage. Kidney Injury Molecule-1 (KIM-1) is a phagocytic receptor upregulated on proximal TECs during acute kidney injury. We have previously shown that renal KIM-1 protects the graft against transplant associated IRI by enabling TECs to clear apoptotic and necrotic cells, and that recognition of necrotic cells by KIM-1 is augmented in the presence of the opsonin, apoptosis inhibitor of macrophages (AIM). Here, we tested whether recombinant AIM (rAIM) could be used to mitigate transplant associated IRI. We administered rAIM or vehicle control to nephrectomised B6 mice transplanted with a single B6 donor kidney. Compared to grafts in vehicle-treated recipients, grafts from rAIM-treated mice exhibited significantly less renal dysfunction, tubular cell death, tissue damage, tubular obstruction, as well as local and systemic inflammation. Both mouse and human rAIM enhanced the clearance of necrotic cells by murine and human TECs, respectively in vitro. These data support testing of rAIM as a potential therapeutic agent to reduce DGF following kidney transplantation.


Subject(s)
Apoptosis Regulatory Proteins/therapeutic use , Delayed Graft Function/drug therapy , Kidney Transplantation/adverse effects , Receptors, Scavenger/therapeutic use , Animals , Apoptosis Regulatory Proteins/administration & dosage , Cells, Cultured , Delayed Graft Function/prevention & control , HEK293 Cells , Humans , Kidney Transplantation/methods , Mice , Mice, Inbred C57BL , Receptors, Scavenger/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
11.
Int J Pharm ; 585: 119535, 2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32534162

ABSTRACT

Induction of apoptosis in tumor cells specifically within the complex tumor microenvironment is highly desirable to kill them efficiently and to enhance the effects of chemotherapy. Second mitochondria-derived activator of caspase (Smac) is a key pro-apoptotic pathway which can be activated with a Smac mimetic peptide. However, in vivo application of peptides is hampered by several limitations such as poor pharmacokinetics, rapid elimination, enzymatic degradation, and insufficient intracellular delivery. In this study, we developed a nanosystem to deliver a Smac peptide to tumor by passive targeting. We first synthesized a chimeric peptide that consists of the 8-mer Smac peptide and a 14-mer cell penetrating peptide (CPP) and then encapsulated the Smac-CPP into polymeric nanoparticles (Smac-CPP-NPs). In vitro, Smac-CPP-NPs were rapidly internalized by 4T1 mammary tumor cells and subsequently released Smac-CPP into the cells, as shown with fluorescence microscopy. Furthermore, Smac-CPP-NPs induced apoptosis in tumor cells, as confirmed with cell viability and caspase 3/7 assays. Interestingly, combination of Smac-CPP-NPs with doxorubicin (dox), a clinically used cytostatic drug, showed combined effects in vitro in 4T1 cells. The effect was significantly better than that of SMAC-CPP-NPs alone as well as empty nanoparticles and dox. In vivo, co-treatment with Smac-CPP-NPs and free dox reduced the tumor growth to 85%. Furthermore, the combination of Smac-CPP-NPs and free dox showed reduced proliferating tumor cells (Ki-67 staining) and increased apoptotic cells (cleaved caspase-3 staining) in tumors. In conclusion, the present study demonstrates that the intracellular delivery of Smac-mimetic peptide using nanoparticle system can be an interesting strategy to attenuate the tumor growth and to potentiate the therapeutic efficacy of chemotherapy in vivo.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Cell-Penetrating Peptides/pharmacology , Doxorubicin/pharmacology , Mitochondrial Proteins/pharmacology , Nanoparticles/chemistry , Animals , Apoptosis Regulatory Proteins/administration & dosage , Cell Death/drug effects , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/pharmacology , Hydrogen-Ion Concentration , Mice , Mitochondrial Proteins/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
12.
J Control Release ; 299: 107-120, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30797866

ABSTRACT

The tumor microenvironment of numerous prevalent cancer types is abundantly infiltrated with tumor-associated macrophages (TAMs). Macrophage mannose receptor (MMR or CD206) expressing TAMs have been shown to be key promoters of tumor progression and major opponents of successful cancer therapy. Therefore, depleting MMR+ TAMs is an interesting approach to synergize with current antitumor therapies. We studied the potential of single-domain antibodies (sdAbs) specific for MMR to target proteins to MMR+ TAMs. Anti-MMR sdAbs were genetically coupled to a reporter protein, mWasabi (wasabi green, WG), generating sdAb "drug" fusion proteins (SFPs), referred to as WG-SFPs. The resulting WG-SFPs were highly efficient in targeting MMR+ macrophages both in vitro and in vivo. As we showed that second mitochondria-derived activator of caspase (SMAC) mimetics modulate MMR+ macrophages, we further coupled the anti-MMR sdAb to an active form of SMAC, referred to as tSMAC. The resulting tSMAC-SFPs were able to bind and upregulate caspase3/7 activity in MMR+ macrophages in vitro. In conclusion, we report the proof-of-concept of an elegant approach to conjugate anti-MMR sdAbs to proteins, which opens new avenues for targeted manipulation of MMR+ tumor-promoting TAMs.


Subject(s)
Drug Delivery Systems , Lectins, C-Type/metabolism , Macrophages/drug effects , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Single-Domain Antibodies/administration & dosage , Animals , Apoptosis Regulatory Proteins/administration & dosage , Apoptosis Regulatory Proteins/pharmacology , Female , HEK293 Cells , Humans , Macrophages/metabolism , Mannose Receptor , Mice, Inbred C57BL , Mitochondrial Proteins/administration & dosage , Mitochondrial Proteins/pharmacology , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Single-Domain Antibodies/pharmacology , Tumor Microenvironment/drug effects
13.
Cancer Sci ; 99(5): 1034-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18294279

ABSTRACT

Although dendritic cells (DC) have been well demonstrated as a strong cellular adjuvant for a tumor vaccine, there are several limitations for clinical application. A protein-based vaccine using a potent adjuvant is an appealing approach for tumor antigen-specific immunotherapy because of their simplicity, safety, efficacy and capacity for repeated administration. CpG-oligodeoxynucleotides (ODN) have been used as adjuvants to stimulate innate and adaptive immune responses for cancer treatment. The authors evaluated the adjuvant effects of CpG-ODN in a vaccine incorporating recombinant fusion protein of the HIV TAT PTD domain and carcinoembryonic antigen (TAT-CEA). Mice vaccinated with TAT-CEA and CpG-ODN (TAT-CEA + CpG) showed enhanced CEA-specific immunity, including cytotoxic T-lymphocytes (CTL) activity and interferon (IFN)-gamma secreting T cells compared with CEA and CpG-ODN (CEA + CpG) or TAT-CEA vaccination alone. Vaccination with TAT-CEA + CpG elicited Th1-based responses, as indicated by the higher ratio of immunoglobulin (Ig)G2a antibody/IgG1 antibodies specific for CEA. The survival rate was significantly increased after vaccination with TAT-CEA + CpG in a tumor model using MC38/CEA2. Furthermore, the TAT-CEA +/- CpG vaccine groups showed similar antitumor immunity to the CEA peptide-pulsed DC (CEA peptide/DC) vaccine groups. These data suggest that coadministration of TAT fusion protein with CpG-ODN may serve as a potential formulation for enhancing antitumor activity.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Apoptosis Regulatory Proteins/therapeutic use , Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Oligodeoxyribonucleotides/therapeutic use , Recombinant Fusion Proteins/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Adjuvants, Immunologic/administration & dosage , Animals , Apoptosis Regulatory Proteins/administration & dosage , Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/administration & dosage , Carcinoembryonic Antigen/therapeutic use , Female , Immunotherapy, Adoptive , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Survivin , T-Lymphocytes, Cytotoxic/immunology , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
14.
Clin Immunol ; 129(2): 341-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18757242

ABSTRACT

T-cell proliferative hyporesponsiveness, a hallmark of paracoccidioidomycosis immune responses, underlies host's failure in controlling fungus spread, being reversible with antifungal treatment. The mechanisms leading to this hypoproliferation are not well known. Since costimulatory molecules have been shown to profoundly regulate T-cell immune responses, we investigated the hypothesis that the determinants of the responder versus tolerant state may be the regulated expression of, or signaling by, costimulatory molecules. Expression of CD80, CD86, CD28, CD152, ICOS and PD-1 costimulatory molecules were examined on T-cells and monocytes harvested from stimulated and unstimulated PBMC cultures of active paracoccidioidomycosis patients and healthy individuals cured of past paracoccidioidomycosis. Stimuli were gp43, the immunodominant component of Paracoccidioides brasiliensis, and a Candida antigen. While CD28 expression, critical for optimal T-cell activation, was comparable between patients and controls, CD152, PD-1 and ICOS, which preferentially deliver negative signaling, were overexpressed on patients' stimulated and unstimulated T-cells. PBMC cultures were carried out in presence of the respective blocking antibodies which, however, failed to restore T-cell proliferation. CD80 and CD86 were equally expressed on patients' and controls' monocytes, but overexpressed on patients' T-cells. Blockade with the respective blocking antibodies on day 4 of the culture also did not restore T-cell proliferation, while, on day 0, differentially inhibited Candida and gp43 responses, suggesting that different antigens require different costimulatory pathways for antigen presentation. Our data favors the hypothesis, raised from other foreign antigen models, that prolonged in vivo antigen exposure leads to an adaptive tolerance T-cell state which is hardly reverted in vitro.


Subject(s)
Antigens, CD/administration & dosage , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , Apoptosis Regulatory Proteins/administration & dosage , B7-1 Antigen/analysis , B7-2 Antigen/analysis , Lymphocyte Activation , Paracoccidioidomycosis/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , CTLA-4 Antigen , Cells, Cultured , Child , Humans , Inducible T-Cell Co-Stimulator Protein , Middle Aged , Monocytes/immunology , Programmed Cell Death 1 Receptor
15.
Cancer Res ; 66(14): 7317-25, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849582

ABSTRACT

The proteasome inhibitor, bortezomib, and the histone deacetylase inhibitor, depsipeptide (FK228), up-regulate tumor death receptors. Therefore, we investigated whether pretreatment of malignant cells with these agents would potentiate natural killer (NK)-mediated tumor killing. NK cells isolated from healthy donors and patients with cancer were expanded in vitro and then tested for cytotoxicity against tumor cell lines before and after exposure to bortezomib or depsipeptide. In 11 of 13 (85%) renal cell carcinoma cell lines and in 16 of 37 (43%) other cancer cell lines, exposure to these drugs significantly increased NK cell-mediated tumor lysis compared with untreated tumor controls (P < 0.001). Furthermore, NK cells expanded from patients with metastatic renal cell carcinoma were significantly more cytotoxic against autologous tumor cells when pretreated with either bortezomib or depsipeptide compared with untreated tumors. Tumors sensitized to NK cell cytotoxicity showed a significant increase in surface expression of DR5 [tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-R2; P < 0.05]; in contrast, surface expression of MHC class I, MIC-A/B, DR4 (TRAIL-R1), and Fas (CD95) did not change. The enhanced susceptibility to NK cell killing was completely abolished by blocking TRAIL on NK cells, and partially abolished by blocking DR5 on tumor cells. These findings show that drug-induced sensitization to TRAIL could be used as a novel strategy to potentiate the anticancer effects of adoptively infused NK cells in patients with cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Boronic Acids/pharmacology , Depsipeptides/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Membrane Glycoproteins/pharmacology , Neoplasms/drug therapy , Pyrazines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins/administration & dosage , Boronic Acids/administration & dosage , Bortezomib , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Depsipeptides/administration & dosage , Drug Synergism , Histone Deacetylase Inhibitors , Humans , Membrane Glycoproteins/administration & dosage , Neoplasms/immunology , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacology , Pyrazines/administration & dosage , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage
16.
Cancer Res ; 66(11): 5867-74, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740726

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (Apo2L/TRAIL) belongs to the TNF family known to transduce their death signals via cell membrane receptors. Because it has been shown that Apo2L/TRAIL induces apoptosis in tumor cells without or little toxicity to normal cells, this cytokine became of special interest for cancer research. Unfortunately, cancer cells are often resistant to Apo2L/TRAIL-induced apoptosis; however, this can be at least partially negotiated by parallel treatment with other substances, such as chemotherapeutic agents. Here, we report that cardiac glycosides, which have been used for the treatment of cardiac failure for many years, sensitize lung cancer cells but not normal human peripheral blood mononuclear cells to Apo2L/TRAIL-induced apoptosis. Sensitization to Apo2L/TRAIL mediated by cardiac glycosides was accompanied by up-regulation of death receptors 4 (DR4) and 5 (DR5) on both RNA and protein levels. The use of small interfering RNA revealed that up-regulation of death receptors is essential for the demonstrated augmentation of apoptosis. Blocking of up-regulation of DR4 and DR5 alone significantly reduced cell death after combined treatment with cardiac glycosides and Apo2L/TRAIL. Combined silencing of DR4 and DR5 abrogated the ability of cardiac glycosides and Apo2L/TRAIL to induce apoptosis in an additive manner. To our knowledge, this is the first demonstration that glycosides up-regulate DR4 and DR5, thereby reverting the resistance of lung cancer cells to Apo2/TRAIL-induced apoptosis. Our data suggest that the combination of Apo2L/TRAIL and cardiac glycosides may be a new interesting anticancer treatment strategy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cardenolides/pharmacology , Cardiac Glycosides/pharmacology , Lung Neoplasms/drug therapy , Membrane Glycoproteins/pharmacology , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/administration & dosage , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cardenolides/administration & dosage , Cardiac Glycosides/administration & dosage , Cell Line, Tumor , Drug Synergism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Glycoproteins/administration & dosage , RNA, Small Interfering/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage , Up-Regulation/drug effects
17.
Cancer Res ; 66(10): 5363-70, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707463

ABSTRACT

Breast cancer is the most common carcinoma that metastasizes to bone. To examine the efficacy of recombinant soluble Apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) against breast cancer growth in bone, we established a mouse model in which MDA-MB-231 human breast cancer cells were transplanted directly into the marrow cavity of the tibiae of athymic nude mice producing osteolytic lesions in the area of injection. All vehicle-treated control animals developed large lesions that established in the marrow cavity, eroded the cortical bone, and invaded the surrounding soft tissue, as assessed by radiography, micro-computed tomography, and histology. In contrast, animals treated with recombinant soluble Apo2L/TRAIL showed significant conservation of the tibiae, with 85% reduction in osteolysis, 90% reduction in tumor burden, and no detectable soft tissue invasion. Tumor cells explanted from Apo2L/TRAIL-treated animals were significantly more resistant to the effects of Apo2L/TRAIL when compared with the cells explanted from the vehicle-treated control animals, suggesting that prolonged treatment with Apo2/TRAIL in vivo selects for a resistant phenotype. However, such resistance was readily reversed when Apo2L/TRAIL was used in combination with clinically relevant chemotherapeutic drugs, including taxol, etoposide, doxorubicin, cisplatin, or the histone deacetylase inhibitor suberoylanilide hydroxamic acid. These studies show for the first time that Apo2L/TRAIL can prevent breast cancer-induced bone destruction and highlight the potential of this ligand for the treatment of metastatic breast cancer in bone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Membrane Glycoproteins/pharmacology , Osteolysis/prevention & control , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/administration & dosage , Breast Neoplasms/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Green Fluorescent Proteins/biosynthesis , Humans , Membrane Glycoproteins/administration & dosage , Mice , Mice, Nude , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage , Xenograft Model Antitumor Assays
18.
Cancer Res ; 66(1): 499-507, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397266

ABSTRACT

Urothelial carcinoma of the bladder accounts for approximately 5% of all cancer deaths in humans. The large majority of tumors are superficial at diagnosis and, after local surgical therapy, have a high rate of local recurrence and progression. Current treatments extend time to recurrence but do not alter disease survival. The objective of the present study was to investigate the tumoricidal potential of combining the apoptosis-inducing protein tumor necrosis factor-related apoptosis inducing ligand (TRAIL) with histone deacetylase inhibitors (HDACi) against TRAIL-resistant bladder tumor cells. Pretreatment with HDACi at nontoxic doses, followed by incubation with TRAIL, resulted in a marked increase in TRAIL-induced apoptosis of T24 cells but showed no significant increase in toxicity to SV40 immortalized normal human uroepithelial cell-1. HDAC inhibition, especially with sodium butyrate and trichostatin A, led to increased TRAIL-R2 gene transcription that correlated with increased TRAIL-R2 surface expression. The increased TRAIL-R2 levels also resulted in accelerated death-inducing signaling complex (DISC) formation, caspase activation, and loss of mitochondrial membrane potential, which all contributed to the increase in tumor cell death. Collectively, these results show the therapeutic potential of combining HDAC inhibition with TRAIL as an alternative treatment for bladder cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Histone Deacetylase Inhibitors , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Urinary Bladder Neoplasms/drug therapy , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/administration & dosage , Apoptosis Regulatory Proteins/genetics , Caspases/metabolism , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondria/drug effects , Mitochondria/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transcription, Genetic , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
19.
Cancer Res ; 66(8): 4309-18, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618756

ABSTRACT

2-Methoxyestradiol is a physiologic metabolite of 17beta-estradiol. This orally active compound can inhibit tumor growth or metastasis in tumor models without inducing any clinical sign of toxicity. Our previous studies indicated that 2-methoxyestradiol-mediated apoptosis involves the disappearance of intact 21-kDa Bid protein, cytochrome c release, and predominant procaspase-3 cleavage. Here, using MIA PaCa-2 cells as a model, we investigated whether this estrogen metabolite induces apoptosis by converging two major pathways: the death receptor-mediated extrinsic and the mitochondrial intrinsic pathway. Exogenous expression of dominant-negative caspase-8 or dominant-negative FADD reverts the effect of 2-methoxyestradiol-mediated cell death. In parallel with this observation, Z-IETD-FMK, a cell permeable irreversible inhibitor of caspase-8, can render significant protection against 2-methoxyestradiol-induced apoptosis. RNase protection assay and cell surface receptor analysis by flow cytometry show the up-regulation of members of death receptor family in 2-methoxyestradiol-exposed pancreatic cancer cells. Our mechanistic studies also implicate that oxidative stress precedes 2-methoxyestradiol-mediated c-Jun NH2-terminal kinase activation, leading to elevated Fas level. Because 2-methoxyestradiol is able to trigger death receptor signaling, we were interested in examining the effects of 2-methoxyestradiol and Fas ligand (FasL)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) together on pancreatic cancer cell death. Interestingly, the endogenous angiogenesis inhibitor 2-methoxyestradiol augments FasL/TRAIL-induced apoptosis in these cells. Moreover, the combination of 2-methoxyestradiol and TRAIL reduces the tumor burden in vivo in MIA PaCa-2 tumor xenograft model by caspase-3 activation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Estradiol/analogs & derivatives , Membrane Glycoproteins/pharmacology , Pancreatic Neoplasms/drug therapy , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology , 2-Methoxyestradiol , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/administration & dosage , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line, Tumor , Drug Synergism , Estradiol/administration & dosage , Estradiol/pharmacology , Humans , Membrane Glycoproteins/administration & dosage , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-jun/metabolism , Receptor Cross-Talk , Signal Transduction/drug effects , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage , Up-Regulation , Xenograft Model Antitumor Assays , fas Receptor/metabolism
20.
Cancer Res ; 66(3): 1740-50, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452234

ABSTRACT

Sulforaphane is a chemopreventive agent present in various cruciferous vegetables, including broccoli. Here, we show that treatment with tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in combination with subtoxic doses of sulforaphane significantly induces rapid apoptosis in TRAIL-resistant hepatoma cells. Neither TNF-alpha- nor Fas-mediated apoptosis was sensitized in hepatoma cells by cotreatment with sulforaphane, suggesting that sulforaphane can selectively sensitize cells to TRAIL-induced apoptosis but not to apoptosis mediated by other death receptors. We found that sulforaphane treatment significantly up-regulated mRNA and protein levels of DR5, a death receptor of TRAIL. This was accompanied by an increase in the generation of reactive oxygen species (ROS). Pretreatment with N-acetyl-l-cysteine and overexpression of catalase inhibited sulforaphane-induced up-regulation of DR5 and almost completely blocked the cotreatment-induced apoptosis. Furthermore, the sulforaphane-mediated sensitization to TRAIL was efficiently reduced by administration of a blocking antibody or small interfering RNAs for DR5. These results collectively indicate that sulforaphane-induced generation of ROS and the subsequent up-regulation of DR5 are critical for triggering and amplifying TRAIL-induced apoptotic signaling. We also found that sulforaphane can sensitize both Bcl-xL- and Bcl-2-overexpressing hepatoma cells to TRAIL-induced apoptosis, indicating that treatment with a combination of TRAIL and sulforaphane may be a safe strategy for treating resistant hepatomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Membrane Glycoproteins/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Thiocyanates/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/administration & dosage , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Drug Synergism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Isothiocyanates , Membrane Glycoproteins/administration & dosage , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Sulfoxides , TNF-Related Apoptosis-Inducing Ligand , Thiocyanates/administration & dosage , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/administration & dosage , Up-Regulation , bcl-X Protein/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL