Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Neurochem Res ; 44(9): 2170-2181, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31420834

ABSTRACT

Acute inhalation of combustion smoke produces long-term neurologic deficits in survivors. To study the mechanisms that contribute to the development of neurologic deficits and identify targets for prevention, we developed a mouse model of acute inhalation of combustion smoke, which supports longitudinal investigation of mechanisms that underlie the smoke induced inimical sequelae in the brain. Using a transgenic mouse engineered to overexpress neuroglobin, a neuroprotective oxygen-binding globin protein, we previously demonstrated that elevated neuroglobin preserves mitochondrial respiration and attenuates formation of oxidative DNA damage in the mouse brain after smoke exposure. In the current study, we show that elevated neuronal neuroglobin attenuates the persistent inflammatory changes induced by smoke exposure in the mouse brain and mitigates concordant smoke-induced long-term neurobehavioral deficits. Specifically, we found that increases in hippocampal density of GFAP and Iba-1 positive cells that are detected post-smoke in wild-type mice are absent in the neuroglobin overexpressing transgenic (Ngb-tg) mice. Similarly, the smoke induced hippocampal myelin depletion is not observed in the Ngb-tg mice. Importantly, elevated neuroglobin alleviates behavioral and memory deficits that develop after acute smoke inhalation in the wild-type mice. Taken together, our findings suggest that the protective effects exerted by neuroglobin in the brains of smoke exposed mice afford protection from long-term neurologic sequelae of acute inhalation of combustion smoke. Our transgenic mouse provides a tool for assessing the potential of elevated neuroglobin as possible strategy for management of smoke inhalation injury.


Subject(s)
Hippocampus/metabolism , Inflammation/metabolism , Neuroglobin/metabolism , Animals , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Gene Expression/drug effects , Hippocampus/pathology , Inflammation/chemically induced , Learning/drug effects , Locomotion/drug effects , Male , Memory/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/drug effects , Neuroglobin/genetics , RNA, Messenger/metabolism , Smoke
2.
Inhal Toxicol ; 29(12-14): 598-610, 2017.
Article in English | MEDLINE | ID: mdl-29405081

ABSTRACT

CONTEXT: Acute inhalation of combustion smoke triggers neurologic sequelae in survivors. Due to the challenges posed by heterogeneity of smoke exposures in humans, mechanistic links between acute smoke inhalation and neuropathologic sequelae have not been systematically investigated. METHODS: Here, using mouse model of acute inhalation of combustion smoke, we studied longitudinal neurobehavioral manifestations of smoke exposures and molecular/cellular changes in the mouse brain. RESULTS: Immunohistochemical analyses at eight months post-smoke, revealed hippocampal astrogliosis and microgliosis accompanied by reduced myelination. Elevated expression of proinflammatory cytokines was also detected. Longitudinal testing in different neurobehavioral paradigms in the course of post-smoke recovery, revealed lasting anxiety-like behavior. The examined paradigms included the open field exploration/anxiety testing at two, four and six months post-smoke, which detected decreases in total distance traveled and time spent in the central arena in the smoke-exposed compared to sham-control mice, suggestive of dampened exploratory activity and increased anxiety-like behavior. In agreement with reduced open field activity, cued fear conditioning test revealed increased freezing in response to conditioned auditory stimulus in mice after acute smoke inhalation. Similarly, elevated plus maze testing demonstrated lesser presence in open arms of the maze, consistent with anxiety-like behavior, for the post-smoke exposure mice. CONCLUSIONS: Taken together, our data demonstrate for the first time persistent neurobehavioral manifestations of acute inhalation of combustion smoke and provide new insights into long-term progression of events initiated by disrupted brain oxygenation that might contribute to lasting adverse sequelae in survivors of smoke inhalation injuries.


Subject(s)
Anxiety/chemically induced , Anxiety/metabolism , Exploratory Behavior/drug effects , Inflammation Mediators/metabolism , Maze Learning/drug effects , Smoke Inhalation Injury/metabolism , Animals , Anxiety/psychology , Exploratory Behavior/physiology , Fear/drug effects , Fear/physiology , Fear/psychology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/psychology , Longitudinal Studies , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Random Allocation , Smoke/adverse effects , Smoke Inhalation Injury/psychology
3.
Inhal Toxicol ; 26(6): 361-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24730682

ABSTRACT

CONTEXT: Acute inhalation of combustion smoke adversely affects brain homeostasis and energy metabolism. We previously showed that overexpressed neuroglobin (Ngb), neuron specific globin protein, attenuates the formation of smoke inhalation-induced oxidative DNA damage, in vivo, in the mouse brain, while others reported protection by Ngb in diverse models of brain injury, mainly involving oxidative stress and hypoxic/ischemic insults. OBJECTIVE: To determine to what extent elevated Ngb ameliorates post smoke-inhalation brain bioenergetics and homeostasis in Ngb overexpressing transgenic mouse. METHODS: Smoke inhalation induced changes in bioenergetics were measured in the wild type and Ngb transgene mouse brain. Modulations of mitochondrial respiration were analyzed using the Seahorse XF24 flux analyzer and changes in cytoplasmic energy metabolism were assessed by measuring enzymatic activities and lactate in the course of post smoke recovery. RESULTS: Cortical mitochondria from Ngb transgene, better maintained ATP synthesis-linked oxygen consumption and unlike wild type mitochondria did not increase futile oxygen consumption feeding the proton leak, reflecting lesser smoke-induced mitochondrial compromise. Measurements revealed lesser reduction of mitochondrial ATP content and lesser compensatory increases in cytosolic energy metabolism, involving pyruvate kinase and lactate dehydrogenase activities as well as cytosolic lactate levels. Additionally, induction of c-Fos, the early response gene and key neuronal stress sensor, was attenuated in Ngb transgene compared to wild type brain after smoke. CONCLUSION: Considered together, these differences reflect lesser perturbations produced by acute inhalation of combustion smoke in the Ngb overexpressing mouse, suggesting that Ngb mitigates mitochondrial dysfunction and neurotoxicity and raises the threshold of smoke inhalation-induced brain injury.


Subject(s)
Brain/metabolism , Globins/physiology , Mitochondria/metabolism , Nerve Tissue Proteins/physiology , Smoke Inhalation Injury/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Genes, fos , Heme Oxygenase-1/analysis , L-Lactate Dehydrogenase/metabolism , Male , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglobin , Oxygen Consumption , Pyruvate Kinase/metabolism
4.
Am J Physiol Gastrointest Liver Physiol ; 305(2): G139-50, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23681476

ABSTRACT

Pancreatitis is classified into acute pancreatitis (AP) and chronic pancreatitis (CP). Apelin, a small regulatory peptide, is the endogenous ligand for the APJ receptor. Apelin and APJ are expressed in the pancreas. The aims of this study were to examine whether apelin influences the inflammatory and fibrosis responses to pancreatitis in mice and to identify mechanisms behind apelin's activities. Supramaximal cerulein induction of AP or CP caused significant (P < 0.05) elevations in pancreatic apelin and APJ expression. Levels declined during the recovery phases. In apelin gene-knockout mice with pancreatitis, pancreatic neutrophil invasion and myeloperoxidase activity were enhanced significantly, and apelin treatment suppressed both. Apelin exposure reduced CP-induced elevations of extracellular matrix-associated proteins. Apelin inhibited PDGF-simulated connective tissue growth factor production and proliferation of pancreatic stellate cells (PSCs). Serum granulocyte colony-stimulating factor and keratinocyte cytokine levels were higher in apelin gene-knockout than wild-type mice with pancreatitis. Apelin reduced AP- and CP-induced elevations in pancreatic NF-κB activation. Together, these findings imply that the pancreatic apelin-APJ system functions to curb the inflammatory and fibrosis responses during pancreatitis. Furthermore, findings suggest that apelin reduces inflammation and fibrosis by reducing neutrophil recruitment and PSC activity. Inhibition of neutrophil invasion may be mediated by reduced keratinocyte cytokine and granulocyte colony-stimulating factor secretion. Apelin-induced reductions in PSC proliferation and connective tissue growth factor production are putative mechanisms underlying apelin's inhibition of extracellular matrix production. The apelin-associated changes in NF-κB binding may be linked to apelin's regulation of pancreatic inflammatory and fibrosis responses during pancreatitis.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Pancreatitis/metabolism , Receptors, G-Protein-Coupled/metabolism , Adipokines , Animals , Apelin , Apelin Receptors , Ceruletide/toxicity , Chemokines , Gene Expression Regulation/physiology , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-3/genetics , Interleukin-3/metabolism , Mice , Mice, Knockout , Pancreatitis/chemically induced , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
Nitric Oxide ; 32: 21-8, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23587847

ABSTRACT

Surges of nitric oxide compromise mitochondrial respiration primarily by competitive inhibition of oxygen binding to cytochrome c oxidase (complex IV) and are particularly injurious in neurons, which rely on oxidative phosphorylation for all their energy needs. Here, we show that transgenic overexpression of the neuronal globin protein, neuroglobin, helps diminish protein nitration, preserve mitochondrial function and sustain ATP content of primary cortical neurons challenged by extended nitric oxide exposure. Specifically, in transgenic neurons, elevated neuroglobin curtailed nitric oxide-induced alterations in mitochondrial oxygen consumption rates, including baseline oxygen consumption, consumption coupled with ATP synthesis, proton leak and spare respiratory capacity. Concomitantly, activation of genes involved in sensing and responding to oxidative/nitrosative stress, including the early-immediate c-Fos gene and the phase II antioxidant enzyme, heme oxygenase-1, was diminished in neuroglobin-overexpressing compared to wild-type neurons. Taken together, these differences reflect a lesser insult produced by similar concentrations of nitric oxide in neuroglobin-overexpressing compared to wild-type neurons, suggesting that abundant neuroglobin buffers nitric oxide and raises the threshold of nitric oxide-mediated injury in neurons.


Subject(s)
Globins/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Oxidative Stress/physiology , Analysis of Variance , Animals , Cerebral Cortex/cytology , Globins/biosynthesis , Globins/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroglobin , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction , Up-Regulation
6.
Pigment Cell Melanoma Res ; 34(3): 605-617, 2021 05.
Article in English | MEDLINE | ID: mdl-33124186

ABSTRACT

Major advances in cancer therapy rely on engagement of the patient's immune system and suppression of mechanisms that impede the antitumor immune response. Among the most notable is immune checkpoint blockade (ICB) therapy that releases immune cells from suppression. Although ICB has had significant success particularly in melanoma, it eradicates tumors in subsets of patients and sequencing data across different cancers suggest that tumors with high mutational loads are more likely to respond to ICB. This is consistent with the premise that greater tumoral mutational loads contribute to formation of neoantigens that spur the body's antitumor immune response. Prompted by strong evidence supporting the therapeutic benefits of neoantigens in the context of ICB, we have developed a mouse melanoma combination treatment, where intratumoral administration of DNA-damaging drug transiently activates intrinsic mutagenic DNA damage tolerance pathway and improves success rates of ICB. Using the YUMM1.7 cells melanoma model, we demonstrate that intratumoral delivery of cisplatin activates translesion synthesis DNA polymerases-catalyzed DNA synthesis on damaged DNA, which when coupled with ICB regimen, elicits durable tumor regression. We expect that this new combination protocol affords insights with clinical relevance that will help expand the range of patients who benefit from ICB therapy.


Subject(s)
Cisplatin/pharmacology , DNA Damage , Immune Checkpoint Inhibitors/pharmacology , Melanoma/drug therapy , Animals , Cross-Linking Reagents/pharmacology , Deoxyuridine/analogs & derivatives , Deoxyuridine/pharmacology , Drug Therapy, Combination , Female , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred C57BL
7.
Front Oncol ; 11: 701968, 2021.
Article in English | MEDLINE | ID: mdl-34295826

ABSTRACT

Sequencing data from different types of cancers including melanomas demonstrate that tumors with high mutational loads are more likely to respond to immune checkpoint blockade (ICB) therapies. We have previously shown that low-dose intratumoral injection of the chemotherapeutic DNA damaging drug cisplatin activates intrinsic mutagenic DNA damage tolerance pathway, and when combined with ICB regimen leads to tumor regression in the mouse YUMM1.7 melanoma model. We now report that tumors generated with an in vitro cisplatin-mutagenized YUMM1.7 clone (YUMM1.7-CM) regress in response to ICB, while an identical ICB regimen alone fails to suppress growth of tumors generated with the parental YUMM1.7 cells. Regressing YUMM1.7-CM tumors show greater infiltration of CD8 T lymphocytes, higher granzyme B expression, and higher tumoral cell death. Similarly, ex-vivo, immune cells isolated from YUMM1.7-CM tumors-draining lymph nodes (TDLNs) co-incubated with cultured YUMM1.7-CM cells, eliminate the tumor cells more efficiently than immune cells isolated from TDLNs of YUMM1.7 tumor-bearing mice. Collectively, our findings show that in vitro induced cisplatin mutations potentiate the antitumor immune response and ICB efficacy, akin to tumor regression achieved in the parental YUMM1.7 model by ICB administered in conjunction with intratumoral cisplatin injection. Hence, our data uphold the role of tumoral mutation burden in improving immune surveillance and response to ICB, suggesting a path for expanding the range of patients benefiting from ICB therapy.

8.
Inhal Toxicol ; 22(9): 770-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20429857

ABSTRACT

Combustion smoke contains gases and particulates, which act via hypoxia and cytotoxicity producing mechanisms to injure cells and tissues. While carbon monoxide (CO) is the major toxicant in smoke, its toxicity is exacerbated in the presence of other compounds. Here, we examined modulations of mitochondrial and cytosolic energy metabolism by inhalation of combustion smoke versus CO, in vivo, in the rat brain. Measurements revealed reduced activities of respiratory chain (RC) complexes, with greater inhibition by smoke than equivalent CO in ambient air. In the case of RC complex IV, inhibition by CO and smoke was similar--suggesting that complex IV inhibition is primarily by the action of CO. In contrast, inhibition of complexes I and III was greater by smoke. Increases in cytosolic lactate dehydrogenase and pyruvate kinase activities accompanied inhibition of RC complexes, likely reflecting compensatory increases in cytosolic energy production. Together, the data provide new insights into the mechanisms of smoke inhalation-induced perturbations of brain energetics, which impact neuronal function and contribute to the development of neuropathologies in survivors of exposures to CO and combustion smoke.


Subject(s)
Brain/drug effects , Carbon Monoxide/toxicity , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Mitochondria/drug effects , Smoke Inhalation Injury/etiology , Smoke/adverse effects , Animals , Brain/metabolism , Disease Models, Animal , Electron Transport Chain Complex Proteins/metabolism , Inhalation Exposure , Lipid Peroxidation/drug effects , Longevity/drug effects , Male , Malondialdehyde/metabolism , Mitochondria/enzymology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Smoke Inhalation Injury/metabolism , Subcellular Fractions
9.
FEBS Open Bio ; 10(5): 789-801, 2020 05.
Article in English | MEDLINE | ID: mdl-32134564

ABSTRACT

The hypoxic environment within solid tumors impedes the efficacy of chemotherapeutic treatments. Here, we demonstrate that hypoxia augments the capacity of melanoma cells to withstand cisplatin and doxorubicin cytotoxicity. We show that B16F10 cells derived from spontaneously formed melanoma and YUMM1.7 cells, engineered to recapitulate human-relevant melanoma driver mutations, profoundly differ in their vulnerabilities to cisplatin and doxorubicin. The differences are manifested in magnitude of proliferative arrest and cell death rates, extent of mtDNA depletion, and impairment of mitochondrial respiration. In both models, cytotoxicity is mitigated by hypoxia, which augments glycolytic metabolism. Collectively, the findings implicate metabolic reprogramming in drug evasion and suggest that melanoma tumors with distinct genetic makeup may have differential drug vulnerabilities, highlighting the importance of precision anticancer treatments.


Subject(s)
Cell Respiration/drug effects , Hypoxia/metabolism , Melanoma/metabolism , Apoptosis/physiology , Cell Line, Tumor , Cell Respiration/physiology , Cisplatin/metabolism , Cisplatin/therapeutic use , Cisplatin/toxicity , Doxorubicin/metabolism , Doxorubicin/therapeutic use , Doxorubicin/toxicity , Glycolysis/physiology , Humans , Melanoma/drug therapy , Mitochondria/metabolism
10.
J Neurochem ; 110(6): 1774-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19619136

ABSTRACT

Tissue-specific iron content is tightly regulated to simultaneously satisfy specialized metabolic needs and avoid cytotoxicity. In the brain, disruption of iron homeostasis may occur in acute as well as progressive injuries associated with neuronal dysfunction and death. We hypothesized that adverse effects of disrupted metal homeostasis on brain function may involve impairment of DNA repair processes. Because in the brain, the base excision repair (BER) pathway is central for handling oxidatively damaged DNA, we investigated effects of elevated iron and zinc on key BER enzymes. In vitro DNA repair assays revealed inhibitory effects of metals on BER activities, including the incision of abasic sites, 5'-flap cleavage, gap filling DNA synthesis and ligation. Using the comet assay, we showed that while metals at concentrations which inhibit BER activities in in vitro assays, did not induce direct genomic damage in cultured primary neurons, they significantly delayed repair of genomic DNA damage induced by sublethal exposure to H(2)O(2). Thus, in the brain even a mild transient metal overload, may adversely affect the DNA repair capacity and thereby compromise genomic integrity and initiate long-term deleterious sequelae including neuronal dysfunction and death.


Subject(s)
Cerebral Cortex/cytology , DNA Damage/drug effects , DNA Repair/physiology , Metals/toxicity , Neurons/drug effects , Neurons/enzymology , Oxidative Stress/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Comet Assay/methods , Electrophoretic Mobility Shift Assay/methods , Hydrogen Peroxide/toxicity , Iron/toxicity , Male , Metals/metabolism , Oxidants/toxicity , Rats , Rats, Sprague-Dawley , Zinc/toxicity
11.
Toxicol Appl Pharmacol ; 235(2): 208-15, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19133281

ABSTRACT

Survivors of massive inhalation of combustion smoke endure critical injuries, including lasting neurological complications. We have previously reported that acute inhalation of combustion smoke disrupts the nitric oxide homeostasis in the rat brain. In this study, we extend our findings and report that a 30-minute exposure of awake rats to ambient wood combustion smoke induces protein nitration in the rat hippocampus and that mitochondrial proteins are a sensitive nitration target in this setting. Mitochondria are central to energy metabolism and cellular signaling and are critical to proper cell function. Here, analyses of the mitochondrial proteome showed elevated protein nitration in the course of a 24-hour recovery following exposure to smoke. Mass spectrometry identification of several significantly nitrated mitochondrial proteins revealed diverse functions and involvement in central aspects of mitochondrial physiology. The nitrated proteins include the ubiquitous mitochondrial creatine kinase, F1-ATP synthase alpha subunit, dihydrolipoamide dehydrogenase (E3), succinate dehydrogenase Fp subunit, and voltage-dependent anion channel (VDAC1) protein. Furthermore, acute exposure to combustion smoke significantly compromised the respiratory capacity of hippocampal mitochondria. Importantly, elevated protein nitration and reduced mitochondrial respiration in the hippocampus persisted beyond the time required for restoration of normal oxygen and carboxyhemoglobin blood levels after the cessation of exposure to smoke. Thus, the time frame for intensification of the various smoke-induced effects differs between blood and brain tissues. Taken together, our findings suggest that nitration of essential mitochondrial proteins may contribute to the reduction in mitochondrial respiratory capacity and underlie, in part, the brain pathophysiology after acute inhalation of combustion smoke.


Subject(s)
Hippocampus/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Nitrates/metabolism , Oxygen Consumption/drug effects , Smoke Inhalation Injury/metabolism , Acute Disease , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Glutamic Acid/metabolism , Hippocampus/drug effects , Male , Mass Spectrometry , Mitochondria/drug effects , Mitochondria/enzymology , Nerve Tissue Proteins/genetics , Oxygen Consumption/genetics , Proteomics , Rats , Rats, Sprague-Dawley , Smoke Inhalation Injury/genetics , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Succinates/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
12.
J Neurochem ; 107(3): 734-44, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18752643

ABSTRACT

Oxidative DNA damage incidental to normal respiratory metabolism poses a particular threat to genomes of highly metabolic-long lived cells. We show that post-mitotic brain has capacity to repair oxidatively damaged DNA ends, which are targets of the long patch (LP) base excision repair (BER) subpathway. LP-BER relies, in part, on proteins associated with DNA replication, including proliferating cell nuclear antigen and is inherent to proliferating cells. Nonetheless, repair products are generated with brain extracts, albeit at slow rates, in the case of 5'-DNA ends modeled with tetrahydrofuran (THF). THF at this position is refractory to DNA polymerase beta 5'-deoxyribose 5-phosphate lyase activity and drives repair into the LP-BER subpathway. Comparison of repair of 5'-THF-blocked termini in the post-mitotic rat brain and proliferative intestinal mucosa, revealed that in mucosa, resolution of damaged 5'-termini is accompanied by formation of larger repair products. In contrast, adducts targeted by the single nucleotide BER are proficiently repaired with both extracts. Our findings reveal mechanistic differences in BER processes selective for the brain versus proliferative tissues. The differences highlight the physiological relevance of the recently proposed 'Hit and Run' mechanism of alternating cleavage/synthesis steps, in the proliferating cell nuclear antigen-independent LP-BER process.


Subject(s)
Brain/physiology , DNA Damage/physiology , DNA Polymerase beta/metabolism , DNA Repair/physiology , Proliferating Cell Nuclear Antigen/metabolism , Animals , Blotting, Western , Immunohistochemistry , Intestinal Mucosa/physiology , Male , Mitosis , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
13.
Free Radic Biol Med ; 44(5): 807-14, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18078825

ABSTRACT

The effects of placental insufficiency and preterm birth on neurodevelopment can be modeled in experimental settings of neonatal hypoxia in rodents. Here, rat pups were reared in reduced oxygen (9.5%) for 11 days, starting on postnatal day 3 (P3). This led to a significant reduction in brain and body weight gain in hypoxic pups compared to age-matched normoxia-reared controls, plausibly reflecting an inability to fulfill the energetic needs of normal growth and development. Adaptive processes designed to augment energetic capacity in eukaryotes include stimulation of mitochondrial biogenesis. We show that after 11 days of sustained hypoxia, the levels of nuclear respiratory factor-1 and mitochondrial transcription factor A are elevated and the content of mitochondrial DNA (mtDNA) is greater in the hypoxic P14 pup brain compared to normoxic conditions. Corresponding immunohistochemical analyses reveal increased density of mtDNA in large cortical neurons. In contrast, no changes in mtDNA content are observed in the brain of pups reared for 24 h (P3-P4) under hypoxic conditions. Together, these data suggest that prolonged inadequate oxygenation may trigger a compensatory increase in neuronal mitochondrial DNA content to partially mitigate compromised energy homeostasis and reduced energetic capacity in the developing hypoxic brain.


Subject(s)
Brain/metabolism , Cell Hypoxia/physiology , DNA, Mitochondrial/genetics , Animals , Animals, Newborn , Blotting, Western , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Malondialdehyde/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Oxygen/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
14.
Mech Ageing Dev ; 129(7-8): 475-82, 2008.
Article in English | MEDLINE | ID: mdl-18374390

ABSTRACT

Accumulation of oxidative DNA damage in the human brain has been implicated in etiologies of post-traumatic and age-associated declines in neuronal function. In neurons, because of high metabolic rates and prolonged life span, exposure to free radicals is intense and risk for accumulation of damaged DNA is amplified. While data indicate that the brain is equipped to repair nuclear and mitochondrial DNA, it is unclear whether repair is executed by distinct subsets of the DNA-repair machinery. Likewise, there are no firm assessments of brain capacity for accurate DNA repair under normal and more so compromised conditions. Consequently, the scope of DNA repair in the brain and the impact of resolution of oxidative lesions on neuronal survival and function remain largely unknown. This review considers evidences for brain levels and activities of the base excision repair (BER) pathway in the context of newly available, comprehensive in situ hybridization analyses of genes encoding repair enzymes. These analyses suggest that not all subsets of BER are equally represented in the brain. Because BER is the major repair process for oxidatively damaged DNA, to what extent parsimonious BER may contribute to development of neuronal dysfunction and brain injury under compromised conditions, is discussed.


Subject(s)
Brain/enzymology , DNA Damage , DNA Repair Enzymes/metabolism , DNA Repair , Neurons/enzymology , Animals , Brain/embryology , Brain/growth & development , Brain Injuries/etiology , Brain Injuries/physiopathology , DNA Repair Enzymes/genetics , Humans , Mice , Neurons/physiology , Oxidative Stress , Rats
15.
Mol Neurobiol ; 55(3): 2506-2515, 2018 03.
Article in English | MEDLINE | ID: mdl-28391554

ABSTRACT

In the peripheral nervous system (PNS) in the absence of tight blood barrier, neurons are at increased risk of DNA damage, yet the question of how effectively PNS neurons manage DNA damage remains largely unanswered. Genotoxins in systemic circulation include chemotherapeutic drugs that reach peripheral neurons and damage their DNA. Because neurotoxicity of platinum-based class of chemotherapeutic drugs has been implicated in PNS neuropathies, we utilized an in vitro model of Dorsal Root Ganglia (DRGs) to investigate how peripheral neurons respond to cisplatin that forms intra- and interstrand crosslinks with their DNA. Our data revealed strong transcriptional upregulation of the translesion synthesis DNA polymerase kappa (Pol κ), while expression of other DNA polymerases remained unchanged. DNA Pol κ is involved in bypass synthesis of diverse DNA lesions and considered a vital player in cellular survival under injurious conditions. To assess the impact of Pol κ deficiency on cisplatin-exposed DRG neurons, Pol κ levels were reduced using siRNA. Pol κ targeting siRNA diminished the cisplatin-induced nuclear Pol κ immunoreactivity in DRG neurons and decreased the extent of cisplatin-induced DNA repair synthesis, as reflected in reduced incorporation of thymidine analog into nuclear DNA. Moreover, Pol κ depletion exacerbated global transcriptional suppression induced by cisplatin in DRG neurons. Collectively, these findings provide the first evidence for critical role of Pol κ in DNA damage response in the nervous system and call attention to implications of polymorphisms that modify Pol κ activity, on maintenance of genomic integrity and neuronal function in exogenously challenged PNS.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , DNA Repair/physiology , DNA-Directed DNA Polymerase/biosynthesis , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Animals , Cells, Cultured , DNA Repair/drug effects , Ganglia, Spinal/pathology , Gene Transfer Techniques , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
16.
Free Radic Biol Med ; 121: 9-19, 2018 06.
Article in English | MEDLINE | ID: mdl-29698743

ABSTRACT

Peripheral Nervous System (PNS) neurotoxicity caused by cancer drugs hinders attainment of chemotherapy goals. Due to leakiness of the blood nerve barrier, circulating chemotherapeutic drugs reach PNS neurons and adversely affect their function. Chemotherapeutic drugs are designed to target dividing cancer cells and mechanisms underlying their toxicity in postmitotic neurons remain to be fully clarified. The objective of this work was to elucidate progression of events triggered by antimitotic drugs in postmitotic neurons. For proof of mechanism study, we chose cytarabine (ara-C), an antimetabolite used in treatment of hematological cancers. Ara-C is a cytosine analog that terminates DNA synthesis. To investigate how ara-C affects postmitotic neurons, which replicate mitochondrial but not genomic DNA, we adapted a model of Dorsal Root Ganglion (DRG) neurons. We showed that DNA polymerase ƎĀ³, which is responsible for mtDNA synthesis, is inhibited by ara-C and that sublethal ara-C exposure of DRG neurons leads to reduction in mtDNA content, ROS generation, oxidative mtDNA damage formation, compromised mitochondrial respiration and diminution of NADPH and GSH stores, as well as, activation of the DNA damage response. Hence, it is plausible that in ara-C exposed DRG neurons, ROS amplified by the high mitochondrial content shifts from physiologic to pathologic levels signaling stress to the nucleus. Combined, the findings suggest that ara-C neurotoxicity in DRG neurons originates in mitochondria and that continuous mtDNA synthesis and reliance on oxidative phosphorylation for energy needs sensitize the highly metabolic neurons to injury by mtDNA synthesis terminating cancer drugs.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Cytarabine/toxicity , DNA, Mitochondrial/metabolism , Ganglia, Spinal/pathology , Mitochondria/pathology , Neurotoxicity Syndromes , Animals , Cells, Cultured , DNA Damage , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Signal Transduction
17.
Free Radic Biol Med ; 42(3): 385-93, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17210451

ABSTRACT

Unrepaired or erroneously repaired DNA lesions drive genomic instability and contribute to cellular and organ decline. Since delayed neuropathologies are common in survivors of smoke inhalation injuries, we asked whether the integrity of brain DNA might be compromised by acute exposure to combustion smoke. Although many studies demonstrate that the brain is equipped to repair oxidatively damaged DNA, to date, the capacity for accurate DNA repair under conditions of disrupted oxygenation and oxidative stress has not been defined. We show that DNA adducts detectable by their ability to block PCR amplification form in the rat hippocampus after acute exposure to smoke. To identify the different types of adducts and to dissect their temporal formation and repair profiles in vivo in the brain, we used DNA-modifying enzymes to convert specific adducts into strand breaks prior to PCR amplification. Using this strategy, we detected formation of oxidative DNA adducts early on after smoke inhalation, while mismatched bases emerged at the later recovery times, potentially due to an erroneous DNA repair process. Erroneous repair can be mutagenic and because the initial smoke-induced oxidative damage to DNA is extensive, compromised fidelity of DNA repair may underlie neurotoxicity and contribute to delayed death of hippocampal neurons.


Subject(s)
DNA Adducts/metabolism , DNA Damage , Hippocampus/metabolism , Oxidative Stress , Smoke Inhalation Injury/metabolism , Animals , Cell Death , Hippocampus/pathology , Lipid Peroxidation , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Smoke Inhalation Injury/pathology
18.
FASEB J ; 20(14): 2639-41, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17060400

ABSTRACT

Apelin, a peptide widely expressed in the body, is the endogenous ligand for the APJ receptor. To investigate how the apelin gene is regulated transcriptionally, we cloned and characterized approximately 3000 and approximately 4000 bp 5'-upstream fragments of the rat and human apelin genes. Putative CAAT-like box, but not TATA-box sites were identified. The rat (-207/-1 bp) and human (-100/+74 bp) core promoter sequences contain putative binding sites for upstream stimulatory factor (USF)-1/-2. Mutagenesis and overexpression assays showed that USF up-regulates basal and inducible apelin transcription. EMSA and supershift experiments indicated binding of USF-1/-2 to the rat (-114/-109 bp) and human (-84/-79 bp) apelin promoters. ChIP experiments show that USF is recruited to the putative USF binding site in the human apelin promoter in cultured breast cells. In concert with increased breast apelin expression during pregnancy and lactation in rats, EMSAs demonstrate an elevated binding of pregnant and lactating rat breast nuclear proteins to a consensus USF oligonucleotide. In vivo ChIP assays verified increased USF binding to the apelin promoter in breast of lactating rats. Together, our findings show that USF exerts a stimulatory role in regulation of breast apelin expression during pregnancy and lactation.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Mammary Glands, Animal/metabolism , Upstream Stimulatory Factors/metabolism , 5' Flanking Region , Animals , Apelin , Base Sequence , Binding Sites , Carrier Proteins/genetics , Cell Line , Female , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Protein Binding , Rats , Rats, Sprague-Dawley , Up-Regulation , Upstream Stimulatory Factors/genetics
19.
Regul Pept ; 142(3): 131-7, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17391779

ABSTRACT

Apelin and its receptor, the APJ receptor, are expressed in the gastrointestinal tract. The aims of this study were to examine the effects of sodium dextran sulfate (DSS)-induced experimental colitis in rats and mice and inflammatory bowel disease (IBD) in humans on intestinal apelin production, and the influence of exogenous apelin on colonic epithelial cell proliferation in mice. In rodents with experimental colitis, colonic apelin mRNA levels were elevated during the inflammatory reaction as well as during the tissue repair phase that ensues after DSS withdrawal. Fluctuations in colonic apelin expression were paralleled by similar changes in apelin immunostaining. Apelin immunostaining was increased in the surface epithelium, in epithelial cells along the length of the tubular gland and in the stem cell region at the gland base. In ulcerative colitis (UC) and Crohn's disease patients, apelin immunostaining revealed a pattern of increased intestinal apelin content similar to that observed in rodents with experimental colitis. Administration of synthetic apelin to mice during the recovery phase of DSS-induced colitis stimulated colonic epithelial cell proliferation significantly. Our observations that colonic apelin production is increased during and after DSS exposure indicate that apelin plays multiple roles during the different stages of colitis. Additionally, the stimulatory action of exogenous apelin on colonic epithelial proliferation suggests that the increased apelin production during intestinal recovery stage may contribute to the repair of the intestinal epithelium in experimental rodent models of colitis and in IBD patients.


Subject(s)
Carrier Proteins/biosynthesis , Colitis/metabolism , Colon/metabolism , Inflammatory Bowel Diseases/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Adipokines , Animals , Apelin , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Proliferation/drug effects , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/drug effects , Colon/pathology , Dextran Sulfate/toxicity , Gene Expression , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
20.
Mol Neurobiol ; 54(10): 7883-7895, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27858292

ABSTRACT

Chemotherapy-induced neurotoxicity of peripheral nervous system (PNS) hinders efficacy of cancer treatments. Mechanisms initiating PNS injury by anticancer drugs are incompletely understood delaying development of effective management strategies. To understand events triggered in PNS by cancer drugs, we exposed dorsal root ganglion (DRG) neurons to cisplatin, a drug from platinum-based class of chemotherapeutics frequently implicated in peripheral neuropathies. While cisplatin enters cancer cells and forms cisplatin/DNA crosslinks that block cell proliferation, circulating cisplatin can also reach the PNS and produce crosslinks that impede critical DNA transactions in postmitotic neurons. Cisplatin forms crosslinks with both, nuclear and mitochondrial DNA (mtDNA). Crosslinks are repairable primarily via the nucleotide excision repair (NER) pathway, which is present in nuclei but absent from mitochondrial compartment. Hence, high mitochondrial content and limited shielding by blood nerve barrier make DRG neurons particularly vulnerable to mitochondrial injury by cisplatin. We report that in DRG neurons, cisplatin elevates reactive oxygen species, depletes mtDNA, and impairs mitochondrial respiration, whereas concomitant meclizine supplementation preserves redox balance, attenuates mitochondrial compromise, and augments DNA repair. Meclizine is an antihistamine drug recently implicated in neuroprotection via modulation of energy metabolism. Our data demonstrate that in the mitochondria-rich DRG neurons, meclizine mitigates cisplatin-induced mitochondrial compromise via enhancement of pentose phosphate pathway and repletion of nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione stores. The findings suggest that meclizine-mediated preservation of redox balance sustains mitochondrial respiration and supports execution of cellular processes, including timely removal of cisplatin crosslinks from nuclear DNA, thereby attenuating cisplatin toxicity in DRG neurons. Collectively, the findings reveal potential for pharmacologic modulation of dorsal root ganglion neurons metabolism for protection against toxicity of chemotherapeutic drugs.


Subject(s)
Cisplatin/pharmacology , DNA Damage/drug effects , Ganglia, Spinal/drug effects , Meclizine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Peripheral Nervous System/drug effects , Peripheral Nervous System/metabolism , Peripheral Nervous System Diseases/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL