Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Exp Neurol ; 342: 113755, 2021 08.
Article En | MEDLINE | ID: mdl-33984337

BACKGROUND: RTP801/REDD1 is a stress-regulated protein whose upregulation is necessary and sufficient to trigger neuronal death in in vitro and in vivo models of Parkinson's and Huntington's diseases and is up regulated in compromised neurons in human postmortem brains of both neurodegenerative disorders. Indeed, in both Parkinson's and Huntington's disease mouse models, RTP801 knockdown alleviates motor-learning deficits. RESULTS: We investigated the physiological role of RTP801 in neuronal plasticity and we found RTP801 in rat, mouse and human synapses. The absence of RTP801 enhanced excitatory synaptic transmission in both neuronal cultures and brain slices from RTP801 knock-out (KO) mice. Indeed, RTP801 KO mice showed improved motor learning, which correlated with lower spine density but increased basal filopodia and mushroom spines in the motor cortex layer V. This paralleled with higher levels of synaptosomal GluA1 and TrkB receptors in homogenates derived from KO mice motor cortex, proteins that are associated with synaptic strengthening. CONCLUSIONS: Altogether, these results indicate that RTP801 has an important role modulating neuronal plasticity and motor learning. They will help to understand its role in neurodegenerative disorders where RTP801 levels are detrimentally upregulated.


Adaptor Proteins, Signal Transducing/deficiency , Learning/physiology , Motor Cortex/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Synapses/genetics
2.
J Vis Exp ; (170)2021 04 17.
Article En | MEDLINE | ID: mdl-33938885

The development of heart failure is the most powerful predictor of long-term mortality in patients surviving acute myocardial infarction (MI). There is an unmet clinical need for prevention and therapy of post-myocardial infarction heart failure (post-MI HF). Clinically relevant pig models of post-MI HF are prerequisites for final proof-of-concept studies before entering into clinical trials in drug and medical device development. Here we aimed to characterize a closed-chest porcine model of post-MI HF in adult Göttingen minipigs with long-term follow-up including serial cardiac magnetic resonance imaging (CMRI) and to compare it with the commonly used Landrace pig model. MI was induced by intraluminal balloon occlusion of the left anterior descending coronary artery for 120 min in Göttingen minipigs and for 90 min in Landrace pigs, followed by reperfusion. CMRI was performed to assess cardiac morphology and function at baseline in both breeds and at 3 and 6 months in Göttingen minipigs and at 2 months in Landrace pigs, respectively. Scar sizes were comparable in the two breeds, but MI resulted in a significant decrease of left ventricular ejection fraction (LVEF) only in Göttingen minipigs, while Landrace pigs did not show a reduction of LVEF. Right ventricular (RV) ejection fraction increased in both breeds despite the negligible RV scar sizes. In contrast to the significant increase of left ventricular end-diastolic (LVED) mass in Landrace pigs at 2 months, Göttingen minipigs showed a slight increase in LVED mass only at 6 months. In summary, this is the first characterization of post-MI HF in Göttingen minipigs in comparison to Landrace pigs, showing that the Göttingen minipig model reflects post-MI HF parameters comparable to the human pathology. We conclude that the Göttingen minipig model is superior to the Landrace pig model to study the development of post-MI HF.


Disease Models, Animal , Heart Failure/etiology , Myocardial Infarction/complications , Animals , Coronary Occlusion/diagnostic imaging , Coronary Occlusion/physiopathology , Female , Heart/diagnostic imaging , Heart/physiopathology , Heart Failure/diagnostic imaging , Heart Failure/physiopathology , Magnetic Resonance Imaging , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Myocardial Reperfusion , Myocardial Reperfusion Injury/physiopathology , Swine , Swine, Miniature , Ventricular Function, Left
3.
Mol Vis ; 24: 712-726, 2018.
Article En | MEDLINE | ID: mdl-30429640

Purpose: Elevations in intraocular pressure (IOP) are associated with the development of glaucoma and loss of sight. High transforming growth factor-ß (TGF-ß) 1 levels in the eye's anterior chamber can lead to dysfunctional contractions through RhoA signaling in trabecular meshwork (TM) cells and IOP spikes. Sustained high TGF-ß levels leads to TM fibrosis and sustained increases in IOP. We investigated whether inhibiting RhoA, using a siRNA-mediated RhoA (siRhoA), controls IOP by altering TM expression of fibrosis and contractility-related proteins in a rodent model of glaucoma. Methods: TGF-ß was injected intracamerally twice a week into adult Sprague Dawley rats, and IOP was recorded with tonometry. Animals were euthanized on day 7 and 35 with TM expression of fibrosis and contractility-related proteins, as well as survival of retinal ganglion cells (RGCs) assessed with immunohistochemistry. siRNA against RhoA or enhanced green fluorescent protein (EGFP) was also injected intracamerally into select animals. Successful RhoA knockdown was determined with quantitative reverse transcription polymerase chain reaction (RT-PCR) and immunohistochemistry, and the effects of the knockdown on the parameters above analyzed. Results: TGF-ß caused increased TM contractile proteins and IOP spikes by day 7, sustained increases in IOP from day 15, and TM fibrosis at day 35. siRhoA abolished the transient 7 day IOP rise but not the later sustained IOP increase (due to fibrosis). At 35 days, TGF-ß-related RGC loss was not prevented with siRhoA treatment. Conclusions: We conclude that RhoA signaling mediates the early IOP rise induced by TM cellular changes associated with contractility but not the sustained IOP elevation caused by TM fibrosis. Thus, RhoA therapies offer a clinically relevant opportunity for IOP management, likely through the modulation of TM contractility, but appear to be ineffective in the amelioration of fibrosis.


Glaucoma, Open-Angle/chemically induced , Intraocular Pressure/drug effects , RNA Interference , Trabecular Meshwork/pathology , Transforming Growth Factor beta1/pharmacology , rhoA GTP-Binding Protein/metabolism , Animals , Disease Models, Animal , Fibrosis/chemically induced , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , RNA Interference/physiology , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tonometry, Ocular , Trabecular Meshwork/metabolism
4.
Invest Ophthalmol Vis Sci ; 57(2): 429-43, 2016 Feb.
Article En | MEDLINE | ID: mdl-26848882

PURPOSE: To investigate, using in vivo and in vitro models, retinal ganglion cell (RGC) neuroprotective and axon regenerative effects and underlying mechanisms of siRTP801, a translatable small-interfering RNA (siRNA) targeting the mTOR negative regulator RTP801. METHODS: Adult rats underwent optic nerve (ON) crush (ONC) followed by intravitreal siRTP801 or control siRNA (siEGFP) every 8 days, with Brn3a+ RGC survival, GFAP+ reactive gliosis, and GAP43+ regenerating axons analyzed immunohistochemically 24 days after injury. Retinal cultures, prepared from uninjured animals or 5 days after ONC to activate retinal glia, were treated with siRTP801/controls in the presence/absence of rapamycin and subsequently assessed for RGC survival and neurite outgrowth, RTP801 expression, glial responses, and mTOR activity. Conditioned medium was analyzed for neurotrophin titers by ELISA. RESULTS: Intravitreal siRTP801 enabled 82% RGC survival compared to 45% with siEGFP 24 days after ONC, correlated with greater GAP43+ axon regeneration at 400 to 1200 µm beyond the ONC site, and potentiated the reactive GFAP+ Müller glial response. In culture, siRTP801 had a direct RGC neuroprotective effect, but required GFAP+ activated glia to stimulate neurite elongation. The siRTP801-induced neuroprotection was significantly reduced, but not abolished, by rapamycin. The siRTP801 potentiated the production and release of neurotrophins NGF, NT-3, and BDNF, and prevented downregulation of RGC mTOR activity. CONCLUSIONS: The RTP801 knockdown promoted RGC survival and axon elongation after ONC, without increasing de novo regenerative sprouting. The neuroprotection was predominantly direct, with mTORC1-dependent and -independent components. Enhanced neurite/axon elongation by siRTP801 required the presence of activated retinal glia and was mediated by potentiated secretion of neurotrophic factors.


Axons/physiology , Gene Expression Regulation/physiology , Nerve Regeneration/physiology , RNA, Small Interfering/pharmacology , Repressor Proteins/genetics , Retinal Ganglion Cells/cytology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Survival/physiology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Intravitreal Injections , Male , Nerve Crush , Nerve Growth Factors/metabolism , Optic Nerve Injuries/etiology , Optic Nerve Injuries/prevention & control , Rats , Rats, Wistar , Retinal Ganglion Cells/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transcription Factors , Transfection
5.
PLoS One ; 10(9): e0135388, 2015.
Article En | MEDLINE | ID: mdl-26367124

There are currently no approved medical radiation countermeasures (MRC) to reduce the lethality of high-dose total body ionizing irradiation expected in nuclear emergencies. An ideal MRC would be effective even when administered well after radiation exposure and would counteract the effects of irradiation on the hematopoietic system and gastrointestinal tract that contribute to its lethality. Entolimod is a Toll-like receptor 5 agonist with demonstrated radioprotective/mitigative activity in rodents and radioprotective activity in non-human primates. Here, we report data from several exploratory studies conducted in lethally irradiated non-human primates (rhesus macaques) treated with a single intramuscular injection of entolimod (in the absence of intensive individualized supportive care) administered in a mitigative regimen, 1-48 hours after irradiation. Following exposure to LD50-70/40 of radiation, injection of efficacious doses of entolimod administered as late as 25 hours thereafter reduced the risk of mortality 2-3-fold, providing a statistically significant (P<0.01) absolute survival advantage of 40-60% compared to vehicle treatment. Similar magnitude of survival improvement was also achieved with drug delivered 48 hours after irradiation. Improved survival was accompanied by predominantly significant (P<0.05) effects of entolimod administration on accelerated morphological recovery of hematopoietic and immune system organs, decreased severity and duration of thrombocytopenia, anemia and neutropenia, and increased clonogenic potential of the bone marrow compared to control irradiated animals. Entolimod treatment also led to reduced apoptosis and accelerated crypt regeneration in the gastrointestinal tract. Together, these data indicate that entolimod is a highly promising potential life-saving treatment for victims of radiation disasters.


Acute Radiation Syndrome/drug therapy , Peptides/therapeutic use , Radiation-Protective Agents/therapeutic use , Toll-Like Receptor 5/agonists , Animals , Bone Marrow/drug effects , Bone Marrow/radiation effects , Female , Hematopoiesis , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Macaca mulatta , Male , Peptides/administration & dosage , Peptides/pharmacology , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/pharmacology
6.
Proc Natl Acad Sci U S A ; 111(21): 7849-54, 2014 May 27.
Article En | MEDLINE | ID: mdl-24825887

Uncoupling protein 1 (Ucp1), which is localized in the mitochondrial inner membrane of mammalian brown adipose tissue (BAT), generates heat by uncoupling oxidative phosphorylation. Upon cold exposure or nutritional abundance, sympathetic neurons stimulate BAT to express Ucp1 to induce energy dissipation and thermogenesis. Accordingly, increased Ucp1 expression reduces obesity in mice and is correlated with leanness in humans. Despite this significance, there is currently a limited understanding of how Ucp1 expression is physiologically regulated at the molecular level. Here, we describe the involvement of Sestrin2 and reactive oxygen species (ROS) in regulation of Ucp1 expression. Transgenic overexpression of Sestrin2 in adipose tissues inhibited both basal and cold-induced Ucp1 expression in interscapular BAT, culminating in decreased thermogenesis and increased fat accumulation. Endogenous Sestrin2 is also important for suppressing Ucp1 expression because BAT from Sestrin2(-/-) mice exhibited a highly elevated level of Ucp1 expression. The redox-inactive mutant of Sestrin2 was incapable of regulating Ucp1 expression, suggesting that Sestrin2 inhibits Ucp1 expression primarily through reducing ROS accumulation. Consistently, ROS-suppressing antioxidant chemicals, such as butylated hydroxyanisole and N-acetylcysteine, inhibited cold- or cAMP-induced Ucp1 expression as well. p38 MAPK, a signaling mediator required for cAMP-induced Ucp1 expression, was inhibited by either Sestrin2 overexpression or antioxidant treatments. Taken together, these results suggest that Sestrin2 and antioxidants inhibit Ucp1 expression through suppressing ROS-mediated p38 MAPK activation, implying a critical role of ROS in proper BAT metabolism.


Gene Expression Regulation/physiology , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Nuclear Proteins/physiology , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Adipocytes/physiology , Animals , Azo Compounds , Fatty Acids, Nonesterified/blood , Humans , Immunoblotting , Mice , Mice, Transgenic , Peroxidases , Real-Time Polymerase Chain Reaction , Uncoupling Protein 1
7.
J Appl Physiol (1985) ; 117(3): 246-56, 2014 Aug 01.
Article En | MEDLINE | ID: mdl-24876363

Although aberrant mTORC1 signaling has been well established in models of obesity, little is known about its repressor, REDD1. Therefore, the initial goal of this study was to determine the role of REDD1 on mTORC1 in obese skeletal muscle. REDD1 expression (protein and message) and mTORC1 signaling (S6K1, 4E-BP1, raptor-mTOR association, Rheb GTP) were examined in lean vs. ob/ob and REDD1 wild-type (WT) vs. knockout (KO) mice, under conditions of altered nutrient intake [fasted and fed or diet-induced obesity (10% vs. 60% fat diet)]. Despite higher (P < 0.05) S6K1 and 4E-BP1 phosphorylation, two models of obesity (ob/ob and diet-induced) displayed elevated (P < 0.05) skeletal muscle REDD1 expression compared with lean or low-fat-fed mouse muscle under fasted conditions. The ob/ob mice displayed elevated REDD1 expression (P < 0.05) that coincided with aberrant mTORC1 signaling (hyperactive S6K1, low raptor-mTOR binding, elevated Rheb GTP; P < 0.05) under fasted conditions, compared with the lean, which persisted in a dysregulated fashion under fed conditions. REDD1 KO mice gained limited body mass on a high-fat diet, although S6K1 and 4E-BP1 phosphorylation remained elevated (P < 0.05) in both the low-fat and high-fat-fed KO vs. WT mice. Similarly, the REDD1 KO mouse muscle displayed blunted mTORC1 signaling responses (S6K1 and 4E-BP1, raptor-mTOR binding) and circulating insulin under fed conditions vs. the robust responses (P < 0.05) in the WT fed mouse muscle. These studies suggest that REDD1 in skeletal muscle may serve to limit hyperactive mTORC1, which promotes aberrant mTORC1 signaling responses during altered nutrient states.


Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Body Mass Index , Carrier Proteins/metabolism , Cell Cycle Proteins , Diet, High-Fat/methods , Eukaryotic Initiation Factors , Food , Insulin/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Obesity/metabolism , Obesity/physiopathology , Phosphoproteins/metabolism , Phosphorylation/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/physiology
8.
J Neurosci ; 33(38): 15295-305, 2013 Sep 18.
Article En | MEDLINE | ID: mdl-24048858

Signals that promote myelination must be tightly modulated to adjust myelin thickness to the axonal diameter. In the peripheral nervous system, axonal neuregulin 1 type III promotes myelination by activating erbB2/B3 receptors and the PI3K/AKT/mTOR pathway in Schwann cells. Conversely, PTEN (phosphatase and tensin homolog on chromosome 10) dephosphorylates PtdIns(3,4,5)P3 and negatively regulates the AKT pathway and myelination. Recently, the DLG1/SAP97 scaffolding protein was described to interact with PTEN to enhance PIP3 dephosphorylation. Here we now report that nerves from mice with conditional inactivation of Dlg1 in Schwann cells display only a transient increase in myelin thickness during development, suggesting that DLG1 is a transient negative regulator of myelination. Instead, we identified DDIT4/RTP801/REDD1 as a sustained negative modulator of myelination. We show that DDIT4 is expressed in Schwann cells and its maximum expression level precedes the peak of AKT activation and of DLG1 activity in peripheral nerves. Moreover, loss of DDIT4 expression both in vitro and in vivo in Ddit4-null mice provokes sustained hypermyelination and enhanced mTORC1 activation, thus suggesting that this molecule is a novel negative regulator of PNS myelination.


Gene Expression Regulation/genetics , Myelin Sheath/metabolism , Schwann Cells/physiology , Transcription Factors/physiology , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Discs Large Homolog 1 Protein , Embryo, Mammalian , Ganglia, Spinal/cytology , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Myelin Basic Protein/metabolism , Myelin P0 Protein/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/physiology , Oligonucleotide Array Sequence Analysis , RNA, Messenger , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SAP90-PSD95 Associated Proteins , Schwann Cells/ultrastructure , Sciatic Nerve/metabolism , Sciatic Nerve/ultrastructure , Transcription Factors/deficiency , Transduction, Genetic
9.
J Neurosci ; 33(5): 2205-16, 2013 Jan 30.
Article En | MEDLINE | ID: mdl-23365256

The transcription factor p53 mediates neuronal death in a variety of stress-related and neurodegenerative conditions. The proapoptotic activity of p53 is tightly regulated by the apoptosis-stimulating proteins of p53 (ASPP) family members: ASPP1 and ASPP2. However, whether ASPP1/2 play a role in the regulation of p53-dependent neuronal death in the CNS is currently unknown. To address this, we asked whether ASPP1/2 contribute to the death of retinal ganglion cells (RGCs) using in vivo models of acute optic nerve damage in mice and rats. Here, we show that p53 is activated in RGCs soon after injury and that axotomy-induced RGC death is attenuated in p53 heterozygote and null mice. We demonstrate that ASPP1/2 proteins are abundantly expressed by injured RGCs, and that short interfering (si)RNA-based ASPP1 or ASPP2 knockdown promotes robust RGC survival. Comparative gene expression analysis revealed that siASPP-mediated downregulation of p53-upregulated-modulator-of-apoptosis (PUMA), Fas/CD95, and Noxa depends on p53 transcriptional activity. Furthermore, siRNA against PUMA or Fas/CD95 confers neuroprotection, demonstrating a functional role for these p53 targets in RGC death. Our study demonstrates a novel role for ASPP1 and ASPP2 in the death of RGCs and provides evidence that blockade of the ASPP-p53 pathway is beneficial for central neuron survival after axonal injury.


Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Death/physiology , Retinal Ganglion Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , fas Receptor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Axons/metabolism , Down-Regulation , Female , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , fas Receptor/genetics
10.
Mol Ther ; 21(4): 834-41, 2013 Apr.
Article En | MEDLINE | ID: mdl-23439501

Notch signaling is active during the development of mosaic epithelial sheets and during their turnover and regeneration. After the loss of hair cells in the mosaic sheet of the vestibular sensory epithelium, new hair cells can be spontaneously generated by transdifferentiation of supporting cells. This regenerative process involves downregulation of the Hes5 gene and is known to be limited and incomplete, especially when the lesion is severe. Here, we test whether further downregulation of Hes5 gene accomplished by the use of siRNA after a severe lesion induced by an aminoglycoside in the mouse utricle can enhance the transdifferentiation of supporting cells and lead to the increased production of new hair cells. We demonstrate that Hes5 levels in the utricle decreased after the application of siRNA and that the number of hair cells in these utricles was significantly larger than following control treatment. The data suggest that siRNA technology may be useful for inducing repair and regeneration in the inner ear and that the Notch signaling pathway is a potentially useful target for specific gene expression inhibition.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Repressor Proteins/metabolism , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Female , Mice , Myosin VIIa , Myosins/genetics , Myosins/metabolism , RNA, Small Interfering , Repressor Proteins/genetics
11.
Am J Respir Cell Mol Biol ; 48(1): 87-93, 2013 Jan.
Article En | MEDLINE | ID: mdl-23024063

Key host responses to the stress induced by environmental exposure to cigarette smoke (CS) are responsible for initiating pathogenic effects that may culminate in emphysema development. CS increases lung ceramides, sphingolipids involved in oxidative stress, structural alveolar cell apoptosis, and inhibition of apoptotic cell clearance by alveolar macrophages, leading to the development of emphysema-like pathology. RTP801, a hypoxia and oxidative stress sensor, is also increased by CS, and has been recently implicated in both apoptosis and inflammation. We investigated whether inductions of ceramide and RTP801 are mechanistically linked, and evaluated their relative importance in lung cell apoptosis and airspace enlargement in vivo. As reported, direct lung instillation of either RTP801 expression plasmid or ceramides in mice triggered alveolar cell apoptosis and oxidative stress. RTP801 overexpression up-regulated lung ceramide levels 2.6-fold. In turn, instillation of lung ceramides doubled the lung content of RTP801. Cell sorting after lung tissue dissociation into single-cell suspension showed that ceramide triggers both endothelial and epithelial cell apoptosis in vivo. Interestingly, mice lacking rtp801 were protected against ceramide-induced apoptosis of epithelial type II cells, but not type I or endothelial cells. Furthermore, rtp801-null mice were protected from ceramide-induced alveolar enlargement, and exhibited improved static lung compliance compared with wild-type mice. In conclusion, ceramide and RTP801 participate in alveolar cell apoptosis through a process of mutual up-regulation, which may result in self-amplification loops, leading to alveolar damage.


Apoptosis/physiology , Ceramides/physiology , DNA-Binding Proteins/physiology , Lung/pathology , Lung/physiopathology , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Emphysema/etiology , Emphysema/pathology , Emphysema/physiopathology , Emphysema/prevention & control , Endothelial Cells/pathology , Endothelial Cells/physiology , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , Lung Compliance/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Smoking/adverse effects , Smoking/pathology , Smoking/physiopathology , Transcription Factors/deficiency , Transcription Factors/genetics
12.
Nucleic Acid Ther ; 22(4): 255-64, 2012 Aug.
Article En | MEDLINE | ID: mdl-22913596

We report the toxicological and pharmacokinetic properties of the synthetic, small interfering RNA I5NP following intravenous administration in rodents and nonhuman primates. I5NP is designed to act via the RNA interference (RNAi) pathway to temporarily inhibit expression of the pro-apoptotic protein p53 and is being developed to protect cells from acute ischemia/reperfusion injuries such as acute kidney injury that can occur during major cardiac surgery and delayed graft function that can occur following renal transplantation. Following intravenous administration, I5NP was very rapidly cleared from plasma was distributed predominantly to the kidney, with very low levels in liver and other tissues. Doses of 800 mg/kg I5NP in rodents, and 1,000 mg/kg I5NP in nonhuman primates, were required to elicit adverse effects, which in the monkey were isolated to direct effects on the blood that included a sub-clinical activation of complement and slightly increased clotting times. In the rat, no additional adverse effects were observed with a rat analogue of I5NP, indicating that the effects likely represent class effects of synthetic RNA duplexes rather than toxicity related to the intended pharmacologic activity of I5NP. Taken together, these data support clinical testing of intravenous administration of I5NP for the preservation of renal function following acute ischemia/reperfusion injury.


RNA, Messenger/genetics , RNA, Small Interfering/toxicity , Tumor Suppressor Protein p53/genetics , Administration, Intravenous , Animals , Area Under Curve , Drug Evaluation, Preclinical , Female , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Macaca fascicularis , Male , Metabolic Clearance Rate , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Rats , Rats, Sprague-Dawley , Renal Insufficiency/metabolism , Tissue Distribution
13.
J Pharmacol Exp Ther ; 343(2): 497-508, 2012 Nov.
Article En | MEDLINE | ID: mdl-22837010

Given an ever-increasing risk of nuclear and radiological emergencies, there is a critical need for development of medical radiation countermeasures (MRCs) that are safe, easily administered, and effective in preventing and/or mitigating the potentially lethal tissue damage caused by acute high-dose radiation exposure. Because the efficacy of MRCs for this indication cannot be ethically tested in humans, development of such drugs is guided by the Food and Drug Administration's Animal Efficacy Rule. According to this rule, human efficacious doses can be projected from experimentally established animal efficacious doses based on the equivalence of the drug's effects on efficacy biomarkers in the respective species. Therefore, identification of efficacy biomarkers is critically important for drug development under the Animal Efficacy Rule. CBLB502 is a truncated derivative of the Salmonella flagellin protein that acts by triggering Toll-like receptor 5 (TLR5) signaling and is currently under development as a MRC. Here, we report identification of two cytokines, granulocyte colony-stimulating factor (G-CSF) and interleukin-6 (IL-6), as candidate biomarkers of CBLB502's radioprotective/mitigative efficacy. Induction of both G-CSF and IL-6 by CBLB502 1) is strictly TLR5-dependent, 2) occurs in a CBLB502 dose-dependent manner within its efficacious dose range in both nonirradiated and irradiated mammals, including nonhuman primates, and 3) is critically important for the ability of CBLB502 to rescue irradiated animals from death. After evaluation of CBLB502 effects on G-CSF and IL-6 levels in humans, these biomarkers will be useful for accurate prediction of human efficacious CBLB502 doses, a key step in the development of this prospective radiation countermeasure.


Biomarkers/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-6/metabolism , Peptides/pharmacology , Animals , Area Under Curve , Cytokines/analysis , Cytokines/metabolism , Dogs , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Granulocyte Colony-Stimulating Factor/genetics , Humans , Injections, Intramuscular , Interleukin-6/genetics , Kaplan-Meier Estimate , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Peptides/administration & dosage , Species Specificity , Whole-Body Irradiation/adverse effects
14.
PLoS One ; 7(3): e33044, 2012.
Article En | MEDLINE | ID: mdl-22479357

Bacterial lipoproteins (BLP) induce innate immune responses in mammals by activating heterodimeric receptor complexes containing Toll-like receptor 2 (TLR2). TLR2 signaling results in nuclear factor-kappaB (NF-κB)-dependent upregulation of anti-apoptotic factors, anti-oxidants and cytokines, all of which have been implicated in radiation protection. Here we demonstrate that synthetic lipopeptides (sLP) that mimic the structure of naturally occurring mycoplasmal BLP significantly increase mouse survival following lethal total body irradiation (TBI) when administered between 48 hours before and 24 hours after irradiation. The TBI dose ranges against which sLP are effective indicate that sLP primarily impact the hematopoietic (HP) component of acute radiation syndrome. Indeed, sLP treatment accelerated recovery of bone marrow (BM) and spleen cellularity and ameliorated thrombocytopenia of irradiated mice. sLP did not improve survival of irradiated TLR2-knockout mice, confirming that sLP-mediated radioprotection requires TLR2. However, sLP was radioprotective in chimeric mice containing TLR2-null BM on a wild type background, indicating that radioprotection of the HP system by sLP is, at least in part, indirect and initiated in non-BM cells. sLP injection resulted in strong transient induction of multiple cytokines with known roles in hematopoiesis, including granulocyte colony-stimulating factor (G-CSF), keratinocyte chemoattractant (KC) and interleukin-6 (IL-6). sLP-induced cytokines, particularly G-CSF, are likely mediators of the radioprotective/mitigative activity of sLP. This study illustrates the strong potential of LP-based TLR2 agonists for anti-radiation prophylaxis and therapy in defense and medical scenarios.


Acute Radiation Syndrome/prevention & control , Hematopoietic System/drug effects , Lipopeptides/pharmacology , Toll-Like Receptor 2/agonists , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/metabolism , Animals , Bacterial Proteins/metabolism , Biomimetic Materials/chemical synthesis , Biomimetic Materials/pharmacology , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/radiation effects , Cytokines/blood , Dose-Response Relationship, Radiation , Female , Granulocyte Colony-Stimulating Factor/blood , HEK293 Cells , Hematopoietic System/radiation effects , Humans , Kaplan-Meier Estimate , Lipopeptides/chemical synthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mycoplasma/metabolism , Spleen/drug effects , Spleen/pathology , Spleen/radiation effects , Thrombocytopenia/etiology , Thrombocytopenia/prevention & control , Time Factors , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Whole-Body Irradiation/adverse effects
15.
Radiat Res ; 177(5): 628-42, 2012 May.
Article En | MEDLINE | ID: mdl-22175300

To date, there are no safe and effective drugs available for protection against ionizing radiation damage. Therefore, a great need exists to identify and develop non-toxic agents that will be useful as radioprotectors or postirradiation therapies under a variety of operational scenarios. We have developed a new pharmacological agent, CBLB613 (a naturally occurring Mycoplasma-derived lipopeptide ligand for Toll-like receptor 2/6), as a novel radiation countermeasure. Using CD2F1 mice, we investigated CBLB613 for toxicity, immunogenicity, radioprotection, radiomitigation and pharmacokinetics. We also evaluated CBLB613 for its effects on cytokine induction and radiation-induced cytopenia in unirradiated and irradiated mice. The no-observable-adverse-effect level of CBLB613 was 1.79 mg/kg and 1 mg/kg for single and repeated doses, respectively. CBLB613 significantly protected mice against a lethal dose of (60)Co γ radiation. The dose reduction factor of CBLB613 as a radioprotector was 1.25. CBLB613 also mitigated the effects of (60)Co γ radiation on survival in mice. In both irradiated and unirradiated mice, the drug stimulated induction of interleukin-1ß (IL-1ß), IL-6, IL-10, IL-12, keratinocyte-derived chemokine, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, and tumor necrosis factor-1α. CBLB613 also reduced radiation-induced cytopenia and increased bone marrow cellularity in irradiated mice. Our immunogenicity study demonstrated that CBLB613 is not immunogenic in mice, indicating that it could be developed as a radioprotector and radiomitigator for humans against the potentially lethal effects of radiation exposure.


Cytokines/blood , Gamma Rays/adverse effects , Lipopeptides/therapeutic use , Mycoplasma/chemistry , Pancytopenia/prevention & control , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/therapeutic use , Toll-Like Receptor 2/agonists , Toll-Like Receptor 6/agonists , Animals , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/radiation effects , Cytokines/biosynthesis , Cytokines/genetics , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Administration Schedule , Drug Evaluation, Preclinical , HEK293 Cells/drug effects , HEK293 Cells/radiation effects , Humans , Lipopeptides/immunology , Lipopeptides/pharmacokinetics , Lipopeptides/toxicity , Male , Mice , NF-kappa B/metabolism , Pancytopenia/blood , Pancytopenia/etiology , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/toxicity , Spleen/drug effects , Spleen/pathology , Spleen/radiation effects
16.
Int J Radiat Oncol Biol Phys ; 83(1): 228-34, 2012 May 01.
Article En | MEDLINE | ID: mdl-22000579

PURPOSE: Development of mucositis is a frequent side effect of radiotherapy of patients with head-and-neck cancer. We have recently reported that bacterial flagellin, an agonist of Toll-like receptor 5 (TLR5), can protect rodents and primates from acute radiation syndrome caused by total body irradiation. Here we analyzed the radioprotective efficacy of TLR5 agonist under conditions of local, single dose or fractionated radiation treatment. METHODS AND MATERIALS: Mice received either single-dose (10, 15, 20, or 25 Gy) or fractioned irradiation (cumulative dose up to 30 Gy) of the head-and-neck area with or without subcutaneous injection of pharmacologically optimized flagellin, CBLB502, 30 min before irradiation. RESULTS: CBLB502 significantly reduced the severity of dermatitis and mucositis, accelerated tissue recovery, and reduced the extent of radiation induced weight loss in mice after a single dose of 15 or 20 Gy but not 25 Gy of radiation. CBLB502 was also protective from cumulative doses of 25 and 30 Gy delivered in two (10 + 15 Gy) or three (3 × 10 Gy) fractions, respectively. While providing protection to normal epithelia, CBLB502 did not affect the radiosensitivity of syngeneic squamous carcinoma SCCVII grown orthotopically in mice. Use of CBLB502 also elicited a radiation independent growth inhibitory effect upon TLR5-expressing tumors demonstrated in the mouse xenograft model of human lung adenocarcinoma A549. CONCLUSION: CBLB502 combines properties of supportive care (radiotherapy adjuvant) and anticancer agent, both mediated via activation of TLR5 signaling in the normal tissues or the tumor, respectively.


Head and Neck Neoplasms/radiotherapy , Peptides/therapeutic use , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/therapeutic use , Radiodermatitis/prevention & control , Stomatitis/prevention & control , Toll-Like Receptor 5/agonists , Adenocarcinoma/metabolism , Animals , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Mice , Mouth Mucosa/radiation effects , Radiation Tolerance/drug effects , Stomatitis/etiology , Toll-Like Receptor 5/metabolism , Weight Loss/drug effects , Xenograft Model Antitumor Assays/methods
17.
J Neurotrauma ; 28(6): 1063-76, 2011 Jun.
Article En | MEDLINE | ID: mdl-21443453

RhoA is a key regulator of the actin cytoskeleton that is upregulated after spinal cord injury (SCI). We analyzed different methods for siRNA delivery and developed siRNAs targeting RhoA (siRhoA) for SCI treatment. Cy 3.5-labeled siRNA delivered at the time of SCI yielded fluorescence in several cell types in the injury site. Intraspinal injections of chemically stabilized siRhoA into the spinal cord of injured rats reduced RhoA protein levels after 1 week and improved hindlimb walking over 6 weeks. To explore a less invasive route, we tested intrathecal injection of Cy 3.5-labeled siRNA via lumbar puncture 1 day after SCI, which resulted in robust uptake in the T9-T10 injury site. Lumbar injection of siRhoA 1 day after SCI reduced RhoA mRNA and protein levels 3 days after injection. Although siRhoA treatment did not yield significant improvement in locomotion, it decreased tactile hypersensitivity significantly compared to controls. Histological analysis at 8 weeks showed significant improvement in white matter sparing with siRhoA compared to control siRNA. siRhoA treatment also resulted in less accumulation of ED1+macrophages, increased PKC-γ immunoreactivity in the corticospinal tract rostral to the injury site, and increased serotonergic fiber growth 12 mm caudal to the contusion site. The ability of siRhoA to preserve white matter and promote serotonergic axonal regrowth caudal to the injury site is likely to suppress allodynia. This provides justification for considering clinical development of RhoA inhibitors to treat SCI sub-acutely to reduce allodynia, which occurs frequently in SCI patients.


Genetic Therapy/methods , Hyperalgesia/therapy , RNA, Small Interfering/administration & dosage , Serotonin/physiology , Spinal Cord Injuries/therapy , rhoA GTP-Binding Protein/administration & dosage , Animals , Disease Models, Animal , Female , Hyperalgesia/genetics , Injections, Spinal , Nerve Regeneration/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/genetics , Up-Regulation/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
18.
Cell Transplant ; 19(12): 1659-70, 2010.
Article En | MEDLINE | ID: mdl-20719069

Renal ischemia-reperfusion (I/R) injury, which is unavoidable in renal transplantation, frequently influences both short- and long-term allograft survival. Despite decades of laboratory and clinical investigations, and the advent of renal replacement therapy, the overall mortality rate due to acute tubular injury has changed little. I/R-induced DNA damage results in p53 activation in proximal tubule cells (PTC), leading to their apoptosis. Therefore, we examined the therapeutic effect of temporary p53 inhibition in two rat renal transplantation models on structural and functional aspects of injury using intravital two-photon microscopy. Nephrectomized Sprague-Dawley rats received syngeneic left kidney transplantation either after 40 min of intentional warm ischemia or after combined 5-h cold and 30-min warm ischemia of the graft. Intravenously administrated siRNA for p53 (siP53) has previously been shown to be filtered and reabsorbed by proximal tubular epithelial cells following the warm ischemia/reperfusion injury in a renal clamp model. Here, we showed that it was also taken up by PTC following 5 h of cold ischemia. Compared to saline-treated recipients, treatment with siP53 resulted in conservation of renal function and significantly suppressed the I/R-induced increase in serum creatinine in both kidney transplantation models. Intravital two-photon microscopy revealed that siP53 significantly ameliorated structural and functional damage to the kidney assessed by quantification of tubular cast formation and the number of apoptotic and necrotic tubular cells and by evaluation of blood flow rate. In conclusion, systemic administration of siRNA for p53 is a promising new approach to protect kidneys from I/R injury in renal transplantation.


Kidney Transplantation/adverse effects , Microscopy/methods , RNA, Small Interfering/therapeutic use , Reperfusion Injury/prevention & control , Tumor Suppressor Protein p53/genetics , Animals , Graft Survival , Kidney/blood supply , Kidney/metabolism , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Transplantation, Homologous , Tumor Suppressor Protein p53/antagonists & inhibitors
19.
Nat Med ; 16(7): 767-73, 2010 Jul.
Article En | MEDLINE | ID: mdl-20473305

Rtp801 (also known as Redd1, and encoded by Ddit4), a stress-related protein triggered by adverse environmental conditions, inhibits mammalian target of rapamycin (mTOR) by stabilizing the TSC1-TSC2 inhibitory complex and enhances oxidative stress-dependent cell death. We postulated that Rtp801 acts as a potential amplifying switch in the development of cigarette smoke-induced lung injury, leading to emphysema. Rtp801 mRNA and protein were overexpressed in human emphysematous lungs and in lungs of mice exposed to cigarette smoke. The regulation of Rtp801 expression by cigarette smoke may rely on oxidative stress-dependent activation of the CCAAT response element in its promoter. We also found that Rtp801 was necessary and sufficient for nuclear factor-kappaB (NF-kappaB) activation in cultured cells and, when forcefully expressed in mouse lungs, it promoted NF-kappaB activation, alveolar inflammation, oxidative stress and apoptosis of alveolar septal cells. In contrast, Rtp801 knockout mice were markedly protected against acute cigarette smoke-induced lung injury, partly via increased mTOR signaling, and, when exposed chronically to cigarette smoke, against emphysema. Our data support the notion that Rtp801 may represent a major molecular sensor and mediator of cigarette smoke-induced lung injury.


Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lung/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pulmonary Emphysema/chemically induced , Smoking/adverse effects , Transcription Factors/physiology , Animals , Enzyme Activation , Homeostasis , Humans , Mice , Mice, Knockout , NF-kappa B/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Emphysema/genetics , TOR Serine-Threonine Kinases , Transcription Factors/genetics
20.
Health Phys ; 98(2): 153-9, 2010 Feb.
Article En | MEDLINE | ID: mdl-20065677

There are urgent needs to establish capability to rapidly assess radiation injury in mass casualty and population monitoring scenarios. This study's objective was to evaluate several currently available biomarkers that can provide early diagnostic triage information after radiation exposure. Hematology and blood chemistry measurements were performed on samples derived from a nonhuman primate (Macaca mulatta; n = 8) total-body irradiation (TBI) model (6.5-Gy Co gamma rays at 0.6 Gy min). The results from this study demonstrate: a) time course for changes in C-reactive protein (CRP) (-2 d to 15 d after TBI); b) time-dependent (-2 d, 1-4 d after TBI) changes in blood cell counts [i.e., lymphocytes decrease to 5-8% of pre-study levels at 1 to 4 d after TBI; ratio of neutrophil to lymphocytes increases by 44 +/- 18 (p = 0.016), 12 +/- 4 (p = 0.001), 8 +/- 2 (p = 0.0020), and 5.0 +/- 2 (p = 0.002) fold at 1, 2, 3, and 4 days after TBI, respectively]; and c) 4.5 +/- 0.8 (p = 0.002)-fold increases in serum amylase activity 1 d after TBI. Plasma CRP levels at 1 d after exposure were 22 +/- 13 (p = 0.0005) (females) and 44 +/- 11 (p = 0.0004) (males)-fold elevated above baseline levels. One hundred percent successful separation of samples from exposed macaques (24 h after TBI) vs. samples from the same macaque taken before irradiation using a discriminant analysis based on four biomarkers (i.e., lymphocytes, neutrophils, ratio of neutrophils to lymphocytes, and serum amylase activity) was demonstrated. These results demonstrate the practical use of multiple parameter biomarkers to enhance the discrimination of exposed vs. non-exposed individuals and justify a follow-on rhesus macaque dose-response study.


Biological Assay/methods , Biomarkers/blood , Models, Biological , Radiation Injuries/blood , Radiation Injuries/diagnosis , Radiometry/methods , Animals , Computer Simulation , Female , Humans , Macaca mulatta , Male , Radiation Dosage , Reproducibility of Results , Risk Assessment/methods , Risk Factors , Sensitivity and Specificity
...