Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
ACS Appl Mater Interfaces ; 14(45): 50583-50591, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36322919

ABSTRACT

Protein therapy has been considered to be one of the most direct and safe ways to regulate cell function and treat tumors. However, safe and effective intracellular delivery of protein drugs is still a key challenge. Herein, we developed a tannic acid-assisted biomineralization strategy for the encapsulation and intracellular delivery of protein drugs. RNase A and glucose oxidase (GOD) were choose as the protein drug model. RNase A, GOD, TA, and Mn2+ are mixed in one pot to attain RG@MT, and CaCO3 coating is subsequently carried out to construct RG@MT@C through biomineralization. Once RG@MT@C is endocytosed, the acidic environment of the lysosome will dissolve the protective layer of CaCO3 and produce plenty of CO2 to cause lysosome bursting, ensuring the lysosome escape of the RG@MT@C and thus releasing the generated TA-Mn2+, RNase A, and GOD into the cytoplasm. The released substances would activate starvation therapy, chemodynamic therapy, and protein therapy pathways to ensure a high performance of cancer therapy. Due to simple preparation, low toxicity, and controlled release in the tumor microenvironment, we expect it can realize efficient and nondestructive delivery of protein drugs and meet the needs for precise, high performance of synergistically antitumor therapy in biomedical applications.


Subject(s)
Nanoparticles , Neoplasms , Humans , Tannins/pharmacology , Tannins/therapeutic use , Ribonuclease, Pancreatic/therapeutic use , Pharmaceutical Preparations , Biomineralization , Neoplasms/drug therapy , Glucose Oxidase/therapeutic use , Cell Line, Tumor , Tumor Microenvironment
2.
ACS Appl Mater Interfaces ; 12(49): 54489-54496, 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33253532

ABSTRACT

A protein-based therapy is significantly challenged by the successful delivery of native proteins into the targeted cancer cells. We address this challenge here using an all-sealed divalent aptamer tetrahedral DNA framework (asdTDF) delivery platform, in which the protein drug is encapsulated inside the cavity of the framework stoichiometrically via a reversible chemical bond. The ligase-assisted seal of the nicks results in highly enhanced TDF stability of the against nuclease digestion to effectively protect the therapeutic protein from degradation. In addition, the divalent aptamer sequences incorporated into the framework favor it with a target-specific and efficient delivery capability. Importantly, upon being readily delivered into the targeted cancer cells, endogenous glutathione can trigger the release of the native therapeutic protein from the TDF in a traceless fashion by cleaving the reversible chemical bond, thereby leading to effective apoptosis of the specific cancer cells.


Subject(s)
DNA/chemistry , Nanostructures/chemistry , Proteins/chemistry , Apoptosis/drug effects , Aptamers, Nucleotide/chemistry , Drug Carriers/chemistry , Fluorescent Dyes/chemistry , Glutathione/chemistry , HeLa Cells , Humans , Neoplasms/drug therapy , Proteins/pharmacology , Proteins/therapeutic use , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/pharmacology , Ribonuclease, Pancreatic/therapeutic use
3.
Int J Mol Sci ; 21(2)2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31936482

ABSTRACT

Angiogenin (ANG) is involved in the innate immune system and inflammatory disease. The aim of this study is to evaluate the anti-inflammatory effects of ANG in an endotoxin induced uveitis (EIU) rat model and the pathways involved. EIU rats were treated with balanced salt solution (BSS), a non-functional mutant ANG (mANG), or wild-type ANG (ANG). The integrity of the blood-aqueous barrier was evaluated by the infiltrating cell and protein concentrations in aqueous humor. Histopathology, Western blot, and real-time qRT-PCR of aqueous humor and ocular tissue were performed to analyze inflammatory cytokines and transcription factors. EIU treated with ANG had decreased inflammatory cells and protein concentrations in the anterior chamber. Compared to BSS and mANG, ANG treatment showed reduced expression of IL-1ß, IL-8, TNF-α, and Myd88, while the expression of IL-4 and IL-10 was increased. Western blot of ANG treatment showed decreased expression of IL-6, inducible nitric oxide synthase (iNOS), IL-1ß, TNF-α, and phosphorylated NF-κB and increased expression of IL-10. In conclusion, ANG seems to reduce effectively immune mediated inflammation in the EIU rat model by reducing the expression of proinflammatory cytokines, while increasing the expression of anti-inflammatory cytokines through pathways related to NF-κB. Therefore, ANG shows potential for effectively suppressing immune-inflammatory responses in vivo.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Ribonuclease, Pancreatic/therapeutic use , Uveitis/chemically induced , Uveitis/drug therapy , Animals , Cell Nucleus/metabolism , Cytokines/genetics , Cytokines/metabolism , Endotoxins , Inflammation/drug therapy , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Models, Biological , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Toll-Like Receptor 4/metabolism
4.
Stomatologiia (Mosk) ; 98(1): 34-37, 2019.
Article in Russian | MEDLINE | ID: mdl-30830091

ABSTRACT

The aim of the study was to study the effect of the gel on the basis of recombinant human angiogenin on the rate of regeneration of donor palatal wounds. The study involved 20 patients (8 men and 12 women) aged 32 to 55 years. Patients were divided into two groups: the 1st group is a study group (n=10), whose patients in the postoperative period used a gel based on recombinant human angiogenin, the 2nd group is a control group (n=10) in which a gel based on recombinant human angiogenin was not used. Patients in both study groups underwent vestibuloplasty with simultaneous plasty of the attached keratinized gingiva with a free gingival graft from the area of the hard palate. The operations were carried out at the stage of disclosing dental implants, simultaneously with the installation of healing abatements or 4 weeks before dental implantation. For histological examination, tissue samples were obtained from the region of the edge of the donor's wounds of the palate at the 7th and 14th days after surgery. As a result of the study, significant differences were found in the comparison groups when assessing the processes of inflammation, angiogenesis and epithelization. The local application of the gel containing recombinant human angiogenin resulted in a rapid decrease in the intensity of inflammation in lamina propria mucosae and a significant decrease in the bulk density of cell infiltrates, accelerating regeneration. This is primarily due to the stimulation of the development of epidermal growth factor (EGF), vascular endothelial growth factor (VEGF) and increased blood supply to the affected area, as well as an increase in the proportion of fibroblasts. The most important observation was the increase in the rate of epithelialization of donor wounds of the hard palate.


Subject(s)
Palate , Ribonuclease, Pancreatic , Vascular Endothelial Growth Factor A , Wound Healing , Adult , Female , Gingiva , Humans , Male , Middle Aged , Palate/injuries , Ribonuclease, Pancreatic/therapeutic use
5.
Georgian Med News ; (276): 177-182, 2018 Mar.
Article in Russian | MEDLINE | ID: mdl-29697403

ABSTRACT

The aim of the study was to evaluate the wound healing properties of the recombinant human angiogenin drug in the gel form on the models of planar musculocutaneous wound and a linear wound. The rats were divided into 3 groups: 1st group - control animals, that did not get any treatment of wounds; 2nd group - experimental, where animals' wound surface was treated with recombinant human angiogenin in a gel medicinal form; and the 3rd group - a comparison group, where animals were treated with "Solcoseryl" drug. The resulted morphology data and the time of complete epithelialization of planar wounds suggest that the human angiogenin drug in gel form has a pronounced wound healing activity. The latter surpasses the studied parameters of the reference drug "Solcoseryl", by contributing to the acceleration of healing process of planar musculocutaneous and linear wounds in rats.


Subject(s)
Ribonuclease, Pancreatic/therapeutic use , Wound Healing/drug effects , Wounds and Injuries/drug therapy , Actihaemyl/therapeutic use , Animals , Gels , Humans , Male , Rats , Recombinant Proteins/therapeutic use , Wounds and Injuries/pathology
6.
Nano Lett ; 18(4): 2294-2303, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29547698

ABSTRACT

Protein therapeutics hold increasing interest with the promise of revolutionizing the cancer treatment by virtue of a potent specific activity and reduced adverse effects. Nonetheless, the therapeutic efficacy of anticancer proteins is highly compromised by multiple successive physiological barriers to protein delivery. In addition, concurrent elimination of bulk tumor cells and highly tumorigenic cancer stem-like cells (CSCs) as a promising strategy has been evidenced to significantly improve cancer therapy. Here we show that a hierarchically assembled nanocomposite can self-adaptively transform its particulate property in response to endogenous tumor-associated signals to overcome the sequential barriers and achieve an enhanced antitumor efficacy by killing CSCs and bulk tumor cells synchronously. The nanoassemblies preferentially accumulate in tumors and dissociate under tumor microenvironmental acidity accompanied by the extracellular release of small-sized ribonuclease A (RNase A)-encapsulated nanocapsule (R-rNC) and small-molecule anti-CSC doxycycline (Doc), which exhibit increased tumor penetration and intracellular accumulation. The endocytosed R-rNC rapidly releases RNase A within both CSCs and tumor cells at intracellular reductive conditions, causing cell death by catalyzing RNA degradation, while Doc eradicates CSCs by inhibiting the mitochondrial biogenesis. The hierarchical assemblies show enhanced cytotoxicity on the CSC-enriched MDA-MB-231 mammospheres and an enhanced antitumor efficacy on the xenograft tumor mouse model.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Breast Neoplasms/drug therapy , Delayed-Action Preparations/chemistry , Doxycycline/administration & dosage , Nanostructures/chemistry , Neoplastic Stem Cells/drug effects , Ribonuclease, Pancreatic/administration & dosage , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Doxycycline/pharmacokinetics , Doxycycline/pharmacology , Doxycycline/therapeutic use , Drug Delivery Systems , Female , Humans , Mice , Ribonuclease, Pancreatic/pharmacokinetics , Ribonuclease, Pancreatic/pharmacology , Ribonuclease, Pancreatic/therapeutic use
7.
Neuropharmacology ; 133: 503-511, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29486168

ABSTRACT

Loss-of-function mutations in the angiogenin (ANG) gene have been identified in familial and sporadic ALS patients. Previous work from our group identified human ANG (huANG) to protect motoneurons in vitro, and provided proof-of-concept that daily intraperitoneal (i.p.) huANG injections post-symptom onset increased lifespan and delayed disease progression in SOD1G93A mice. huANG's mechanism of action remains less well understood. Here, we implemented a preclinical in vivo design to validate our previous results, provide pharmacokinetic and protein distribution data after systemic administration, and explore potential pleiotropic activities of huANG in vivo. SOD1G93A mice (n = 45) and non-transgenic controls (n = 31) were sex- age- and litter-matched according to the 2010 European ALS/MND group guidelines, and treated with huANG (1 µg, i.p., 3 times/week) or vehicle from 90 days on. huANG treatment increased survival and delayed motor dysfunction as assessed by rotarod in SOD1G93A mice. Increased huANG serum levels were detectable 2 and 24 h after i.p. injection equally in transgenic and non-transgenic mice. Exogenous huANG localized to spinal cord astrocytes, supporting a glia-mediated, paracrine mechanism of action; uptake into endothelial cells was also observed. 1 µg huANG or vehicle were administered from 90 to 115 days of age for histological analysis. Vehicle-treated SOD1G93A mice showed decreased motoneuron numbers and vascular length per ventral horn area, while huANG treatment resulted in improved vascular network maintenance and motoneuron survival. Our data suggest huANG represents a new class of pleiotropic ALS therapeutic that acts on the spinal cord vasculature and glia to delay motoneuron degeneration and disease progression.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Angiogenesis Inducing Agents/therapeutic use , Ribonuclease, Pancreatic/therapeutic use , Age Factors , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/complications , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/drug effects , Motor Neurons/pathology , Movement Disorders/drug therapy , Movement Disorders/etiology , Ribonuclease, Pancreatic/blood , Rotarod Performance Test , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Survival Analysis , Time Factors
8.
Biomed Res Int ; 2017: 9390565, 2017.
Article in English | MEDLINE | ID: mdl-29259991

ABSTRACT

OBJECTIVE: This study aimed to develop a gene delivery system using ultrasound-targeted microbubbles destruction (UTMD) combined with nuclear localization signal (NLS) and investigate its efficacy and safety for therapeutic angiogenesis in canine myocardial infarction (MI) model. METHODS: Fifty MI dogs were randomly divided into 5 groups and transfected with Ang-1 gene plasmid: (i) group A: only injection of microbubbles and Ang-1 plasmid; (ii) group B: only UTMD mediated gene transfection; (iii) group C: UTMD combined with classical NLS mediated gene transfection; (iv) group D: UTMD combined with mutational NLS mediated transfection; and (v) group E: UTMD combined with classical NLS in the presence of a nucleus transport blocker. The mRNA and protein expression of Ang-1 gene, microvessel density (MVD) cardiac troponin I (cTnI), and cardiac function were determined after transfection. RESULTS: The expression of mRNA and protein of Ang-1 gene in group C was significantly higher than that of the other groups (all P < 0.01). The MVD of group C was 10.2-fold of group A and 8.1-fold of group E (P < 0.01). The cardiac function in group C was significant improvement without cTnI rising. CONCLUSIONS: The gene delivery system composed of UTMD and NLS is efficient and safe.


Subject(s)
Dog Diseases/therapy , Gene Transfer Techniques , Microbubbles/therapeutic use , Myocardial Infarction/therapy , Ribonuclease, Pancreatic/therapeutic use , Animals , Dog Diseases/genetics , Dog Diseases/physiopathology , Dogs , Gene Expression Regulation , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , RNA, Messenger , Ribonuclease, Pancreatic/genetics , Troponin I/genetics , Ultrasonic Waves
9.
Oncotarget ; 7(52): 87016-87030, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27894092

ABSTRACT

Breast cancer is the most common cancer in women worldwide. A new promising anti-cancer therapy involves the use of monoclonal antibodies specific for target tumor-associated antigens (TAAs). A TAA of interest for immunotherapy of Triple Negative Breast Cancer (TNBC) is nucleolin (NCL), a multifunctional protein, selectively expressed on the surface of cancer cells, which regulates the biogenesis of specific microRNAs (miRNAs) involved in tumor development and drug-resistance. We previously isolated a novel human anti-NCL scFv, called 4LB5, that is endowed with selective anti-tumor effects. Here we report the construction and characterization of a novel immunoRNase constituted by 4LB5 and a human pancreatic RNase (HP-RNase) called "4LB5-HP-RNase". This immunoRNase retains both the enzymatic activity of human pancreatic RNase and the specific binding of the parental scFv to a panel of surface NCL-positive breast cancer cells. Notably, 4LB5-HP-RNase dramatically and selectively reduced the viability and proliferation of NCL-positive tumor cells in vitro and in vivo. Specifically, it induced apoptosis and reduced the levels of the tumorigenic miRNAs miR-21, -221 and -222. Thus, this novel immunoagent could be a valuable tool for the treatment of TNBC patients ineligible for currently available targeted treatments.


Subject(s)
Phosphoproteins/immunology , RNA-Binding Proteins/immunology , Ribonuclease, Pancreatic/therapeutic use , Single-Chain Antibodies/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Vesicles/physiology , Female , Humans , Mice , MicroRNAs/analysis , Phosphoproteins/analysis , RNA-Binding Proteins/analysis , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , Nucleolin
10.
Zhonghua Shao Shang Za Zhi ; 31(6): 473-5, 2015 Dec.
Article in Chinese | MEDLINE | ID: mdl-26837255

ABSTRACT

Angiogenin, as a member of the ribonuclease superfamily, is an angiogenic protein. The angiogenic ability of angiogenin plays an important role in many physical and pathological processes. Angiogenin can induce endothelial cell migration, proliferation, tubulation, as well as inhibition of cellular apoptosis. Angiogenin can be used to modulate the angiogenetic process of tissue engineered constructions via local delivery. Furthermore, angiogenin can also be regarded as a biomarker for diagnostic evaluation of malignancy, or as a target for cancer therapy. This paper presents a comprehensive overview of the angiogenic mechanisms of angiogenin, as well as its potential application in the process of wound healing and treatment of ischemic diseases and malignancy.


Subject(s)
Cell Movement , Neovascularization, Pathologic , Ribonuclease, Pancreatic/therapeutic use , Humans , Neovascularization, Physiologic
11.
Angew Chem Int Ed Engl ; 53(49): 13444-8, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25287050

ABSTRACT

Herein we report a convenient chemical approach to reversibly modulate protein (RNase A) function and develop a protein that is responsive to reactive oxygen species (ROS) for targeted cancer therapy. The conjugation of RNase A with 4-nitrophenyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) benzyl carbonate (NBC) blocks protein lysine and temporarily deactivates the protein. However, the treatment of RNase A-NBC with hydrogen peroxide (one major intracellular ROS) efficiently cleaves the NBC conjugation and restores the RNase A activity. Thus, RNase A-NBC can be reactivated inside tumor cells by high levels of intracellular ROS, thereby restoring the cytotoxicity of RNase A for cancer therapy. Due to higher ROS levels inside tumor cells compared to healthy cells, and the resulting different levels of RNase A-NBC reactivation, RNase A-NBC shows a significant specific cytotoxicity against tumor cells.


Subject(s)
Benzyl Compounds/chemistry , Carbonates/chemistry , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/therapeutic use , Benzyl Compounds/metabolism , Carbonates/metabolism , Cell Line, Tumor , Enzyme Therapy , Humans , Neoplasms/enzymology , Neoplasms/metabolism , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/pharmacology
12.
Angew Chem Int Ed Engl ; 53(11): 2893-8, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24519972

ABSTRACT

An efficient and safe method to deliver active proteins into the cytosol of targeted cells is highly desirable to advance protein-based therapeutics. A novel protein delivery platform has been created by combinatorial design of cationic lipid-like materials (termed "lipidoids"), coupled with a reversible chemical protein engineering approach. Using ribonuclease A (RNase A) and saporin as two representative cytotoxic proteins, the combinatorial lipidoids efficiently deliver proteins into cancer cells and inhibit cell proliferation. A study of the structure-function relationship reveals that the electrostatic and hydrophobic interactions between the lipidoids and the protein play a vital role in the formation of protein-lipidoid nanocomplexes and intracellular delivery. A representative lipidoid (EC16-1) protein nanoparticle formulation inhibits cell proliferation in vitro and suppresses tumor growth in a murine breast cancer model.


Subject(s)
Antineoplastic Agents/administration & dosage , Lipids/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Aconitic Acid/analogs & derivatives , Aconitic Acid/chemistry , Alkanes/chemistry , Amines/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , Humans , Mice , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/therapeutic use , Ribonuclease, Pancreatic/toxicity , Ribosome Inactivating Proteins, Type 1/metabolism , Ribosome Inactivating Proteins, Type 1/therapeutic use , Ribosome Inactivating Proteins, Type 1/toxicity , Saporins
13.
Khirurgiia (Mosk) ; (2): 25-9, 2013.
Article in Russian | MEDLINE | ID: mdl-23503379

ABSTRACT

The efficacy and safety of gene-engineering recombinant constructions with endothelial growth factor gene and angiogenin for the treatment of the chronic lower limb ischemia were studied. 134 patients were included in prospective controlled study. The main group, who received both traditional treatment and genetic therapy, consisted of 74 patients. The rest 60 patients were included into the control group. Of 74 patients from the main group, genetic therapy was used together with conservative means in 39 patients and with reconstructive vascular operations in 35 patients. The gene-engineering angiogenesis stimulation therapy proved to be effective and safe. The combination of angiogenesis genetic stimulation with reconstructive vascular surgery demonstrated significantly better results, then monotherapy.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Genetic Therapy/methods , Ischemia/therapy , Leg/blood supply , Ribonuclease, Pancreatic/therapeutic use , Vascular Surgical Procedures , Adult , Aged , Aged, 80 and over , Chronic Disease , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neovascularization, Pathologic/prevention & control , Prospective Studies , Treatment Outcome
14.
Bioorg Med Chem Lett ; 21(9): 2756-8, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21144746

ABSTRACT

Variants of bovine pancreatic ribonuclease (RNase A) engineered to evade the endogenous ribonuclease inhibitor protein (RI) are toxic to human cancer cells. Increasing the basicity of these variants facilitates their entry into the cytosol and thus increases their cytotoxicity. The installation of additional positive charge also has the deleterious consequence of decreasing ribonucleolytic activity or conformational stability. Here, we report that the same benefit can be availed by co-treating cells with a cationic dendrimer. We find that adding the generation 2 poly(amidoamine) dendrimer in trans increases the cytotoxicity of RI-evasive RNase A variants without decreasing their activity or stability. The increased cytotoxicity is not due to increased RI-evasion or cellular internalization, but likely results from improved translocation into the cytosol after endocytosis. These data indicate that co-treatment with highly cationic molecules could enhance the efficacy of ribonucleases as chemotherapeutic agents.


Subject(s)
Dendrimers/chemistry , Polyamines/chemistry , Ribonuclease, Pancreatic/chemistry , Animals , Cations , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Dendrimers/toxicity , Drug Therapy, Combination , Genetic Variation , Humans , Inhibitory Concentration 50 , K562 Cells , Models, Molecular , Neoplasms/drug therapy , Polyamines/toxicity , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/therapeutic use
15.
J Neurochem ; 116(3): 334-41, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21091473

ABSTRACT

We previously observed marked down-regulation of the mRNA for angiogenin, a potent inducer of neovascularization, in a mouse model of Parkinson's disease (PD) based on over-expression of alpha-synuclein. Angiogenin has also been recently implicated in the pathogenesis of amyotrophic lateral sclerosis. In this study, we confirmed that mouse angiogenin-1 protein is dramatically reduced in this transgenic alpha-synuclein mouse model of PD, and examined the effect of angiogenin in cellular models of PD. We found that endogenous angiogenin is present in two dopamine-producing neuroblastoma cell lines, SH-SY5Y and M17, and that exogenous angiogenin is taken up by these cells and leads to phosphorylation of Akt. Applied angiogenin protects against the cell death induced by the neurotoxins 1-methyl-4-phenylpyridinium and rotenone and reduces the activation of caspase 3. Together our data supports the importance of angiogenin in protecting against dopaminergic neuronal cell death and suggests its potential as a therapy for PD.


Subject(s)
Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Ribonuclease, Pancreatic/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Cytoprotection/drug effects , Cytoprotection/physiology , Disease Models, Animal , Dopamine/biosynthesis , Dopamine/physiology , Humans , Mice , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/drug therapy , Nerve Degeneration/genetics , Nerve Degeneration/prevention & control , Neuroblastoma/enzymology , Neuroblastoma/pathology , Neuroblastoma/therapy , Neuroprotective Agents/metabolism , Oncogene Protein v-akt/metabolism , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/therapeutic use
17.
Curr Pharm Des ; 15(23): 2665-75, 2009.
Article in English | MEDLINE | ID: mdl-19689337

ABSTRACT

Ribonucleases degrade RNA, now considered an important drug target. The parent member of this protein superfamily is bovine pancreatic RNase A that functions as a digestive enzyme. Other physiological roles and activities have been ascribed to more recently discovered members of this superfamily. Angiogenin was isolated by following angiogenic activity from cell culture media conditioned by colon cancer cells. ONCONASE kills tumor cells in vitro and in vivo and has advanced to a phase IIIb confirmatory clinical trial for the treatment of unresectable malignant mesothelioma. All three of these RNA degrading enzymes have been used to generate immunoRNases; chemical conjugates and ligand-RNase fusion proteins, for cancer therapy. The properties of each of these RNases are described along with the increasingly sophisticated construction of recombinant immunoRNases. The advantages of using RNase as an antibody payload is compared to using plant or bacterial toxins in the construction of immunotoxins, a related strategy for specifically killing malignant cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Discovery/methods , Ribonucleases/therapeutic use , Animals , Humans , Immunotoxins/therapeutic use , Models, Molecular , Recombinant Proteins/biosynthesis , Recombinant Proteins/therapeutic use , Ribonuclease, Pancreatic/therapeutic use , Ribonucleases/immunology , Ribonucleases/physiology
18.
Expert Opin Biol Ther ; 9(1): 79-95, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19063695

ABSTRACT

Immunotoxins combining antibody specificity with bacterial or plant toxins are limited by their strong immunogenicity and non-specific toxicity. Ribonucleases of the RNase A protein superfamily provide a solution to address these issues because they show potent antineoplastic activity on cell internalization but do not show appreciable immunogenicity or non-specific toxicity. Their therapeutic value is demonstrated by RNase derived from the frog (Rana pipiens), Onconase (ONC, Alfacell, Inc., New Jersey, USA), the first and only RNase being evaluated in clinical trials at present. Conjugation or fusion of RNases to tumor specific antibodies is a promising approach to further boost tumor cell killing of these compounds. This review focuses on 'targeted RNases/ImmunoRNases' as promising novel anticancer therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Ribonucleases/therapeutic use , Animals , Humans , Immunotoxins/therapeutic use , Ki-1 Antigen/immunology , Prodrugs/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Ribonuclease, Pancreatic/therapeutic use , Sialic Acid Binding Ig-like Lectin 2/immunology
19.
J Neurosci ; 28(52): 14056-61, 2008 Dec 24.
Article in English | MEDLINE | ID: mdl-19109488

ABSTRACT

Mutations in the hypoxia-inducible factor angiogenin (ANG) have been identified in Amyotrophic Lateral Sclerosis (ALS) patients, but the potential role of ANG in ALS pathogenesis was undetermined. Here we show that angiogenin promotes motoneuron survival both in vitro and in vivo. Angiogenin protected cultured motoneurons against excitotoxic injury in a PI-3-kinase/Akt kinase-dependent manner, whereas knock-down of angiogenin potentiated excitotoxic motoneuron death. Expression of wild-type ANG protected against endoplasmic reticulum (ER) stress-induced and trophic-factor-withdrawal-induced cell death in vitro, whereas the ALS-associated ANG mutant K40I exerted no protective activity and failed to activate Akt-1. In SOD1(G93A) mice angiogenin delivery increased lifespan and motoneuron survival, restored the disease-associated decrease in Akt-1 survival signaling, and reversed a pathophysiological increase in ICAM-1 expression. Our data demonstrate that angiogenin is a key factor in the control of motoneuron survival.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Motor Neurons/drug effects , Neuroprotective Agents/pharmacology , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/pharmacology , Androstadienes/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Female , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Insulin-Like Growth Factor I/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed/methods , Oncogene Protein v-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/therapeutic use , Spinal Cord/cytology , Superoxide Dismutase/genetics , Time Factors , Transfection/methods , Tunicamycin/pharmacology , Wortmannin , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...