Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 132(17)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36047496

RESUMO

Cancer-related cognitive impairment (CRCI) is a major neurotoxicity affecting more than 50% of cancer survivors. The underpinning mechanisms are mostly unknown, and there are no FDA-approved interventions. Sphingolipidomic analysis of mouse prefrontal cortex and hippocampus, key sites of cognitive function, revealed that cisplatin increased levels of the potent signaling molecule sphingosine-1-phosphate (S1P) and led to cognitive impairment. At the biochemical level, S1P induced mitochondrial dysfunction, activation of NOD-, LRR-, and pyrin domain-containing protein 3 inflammasomes, and increased IL-1ß formation. These events were attenuated by systemic administration of the functional S1P receptor 1 (S1PR1) antagonist FTY720, which also attenuated cognitive impairment without adversely affecting locomotor activity. Similar attenuation was observed with ozanimod, another FDA-approved functional S1PR1 antagonist. Mice with astrocyte-specific deletion of S1pr1 lost their ability to respond to FTY720, implicating involvement of astrocytic S1PR1. Remarkably, our pharmacological and genetic approaches, coupled with computational modeling studies, revealed that cisplatin increased S1P production by activating TLR4. Collectively, our results identify the molecular mechanisms engaged by the S1P/S1PR1 axis in CRCI and establish S1PR1 antagonism as an approach to target CRCI with therapeutics that have fast-track clinical application.


Assuntos
Disfunção Cognitiva , Cloridrato de Fingolimode , Animais , Sistema Nervoso Central/metabolismo , Cisplatino/efeitos adversos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/genética , Cloridrato de Fingolimode/farmacologia , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/genética
2.
Neurosci Lett ; 760: 136087, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34182057

RESUMO

Chemotherapy-induced peripheral neuropathy (CIPN) is a somatosensory axonopathy in cancer patients receiving any of a variety of widely-use antitumor agents. CIPN can lead to long-lasting neuropathic pain that limits the dose or length of otherwise life-saving cancer therapy. Accumulating evidence over the last two decades indicates that many chemotherapeutic agents cause mitochondrial injury in the peripheral sensory nerves by disrupting mitochondrial structure and bioenergetics, increasing nitro-oxidative stress and altering mitochondrial transport, fission, fusion and mitophagy. The accumulation of abnormal and dysfunctional mitochondria in sensory neurons are linked to axonal growth defects resulting in the loss of intraepidermal nerve fibers in the hands and feet, increased spontaneous discharge and the sensitization of peripheral sensory neurons that provoke and promote changes in the central nervous system that establish a chronic neuropathic pain state. This has led to the propose mitotoxicity theory of CIPN. Strategies that improve mitochondrial function have shown success in preventing and reversing CIPN in pre-clinical animal models and have begun to show some progress toward translation to the clinic. In this review, we will review the evidence for, the causes and effects of and current strategies to target mitochondrial dysfunction in CIPN.


Assuntos
Antineoplásicos/efeitos adversos , Mitocôndrias/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neuralgia/induzido quimicamente , Células Receptoras Sensoriais/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Modelos Animais de Doenças , Humanos , Mitocôndrias/patologia , Neuralgia/patologia , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/patologia
3.
J Neuroinflammation ; 17(1): 339, 2020 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-33183330

RESUMO

BACKGROUND: Traumatic brain injury (TBI) is a common pathological condition that presently lacks a specific pharmacological treatment. Adenosine levels rise following TBI, which is thought to be neuroprotective against secondary brain injury. Evidence from stroke and inflammatory disease models suggests that adenosine signaling through the G protein-coupled A3 adenosine receptor (A3AR) can provide antiinflammatory and neuroprotective effects. However, the role of A3AR in TBI has not been investigated. METHODS: Using the selective A3AR agonist, MRS5980, we evaluated the effects of A3AR activation on the pathological outcomes and cognitive function in CD1 male mouse models of TBI. RESULTS: When measured 24 h after controlled cortical impact (CCI) TBI, male mice treated with intraperitoneal injections of MRS5980 (1 mg/kg) had reduced secondary tissue injury and brain infarction than vehicle-treated mice with TBI. These effects were associated with attenuated neuroinflammation marked by reduced activation of nuclear factor of kappa light polypeptide gene enhancer in B cells (NFκB) and MAPK (p38 and extracellular signal-regulated kinase (ERK)) pathways and downstream NOD-like receptor pyrin domain-containing 3 inflammasome activation. MRS5980 also attenuated TBI-induced CD4+ and CD8+ T cell influx. Moreover, when measured 4-5 weeks after closed head weight-drop TBI, male mice treated with MRS5980 (1 mg/kg) performed significantly better in novel object-placement retention tests (NOPRT) and T maze trials than untreated mice with TBI without altered locomotor activity or increased anxiety. CONCLUSION: Our results provide support for the beneficial effects of small molecule A3AR agonists to mitigate secondary tissue injury and cognitive impairment following TBI.


Assuntos
Agonistas do Receptor A3 de Adenosina/administração & dosagem , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Transtornos Neurocognitivos/tratamento farmacológico , Transtornos Neurocognitivos/metabolismo , Receptor A3 de Adenosina/metabolismo , Animais , Lesões Encefálicas Traumáticas/patologia , Sistemas de Liberação de Medicamentos/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Neurocognitivos/patologia
4.
J Pharmacol Exp Ther ; 375(2): 367-375, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32913007

RESUMO

Neuropathic pain is a debilitating public health concern for which novel non-narcotic therapeutic targets are desperately needed. Using unbiased transcriptomic screening of the dorsal horn spinal cord after nerve injury we have identified that Gpr183 (Epstein-Barr virus-induced gene 2) is upregulated after chronic constriction injury (CCI) in rats. GPR183 is a chemotactic receptor known for its role in the maturation of B cells, and the endogenous ligand is the oxysterol 7α,25-dihydroxycholesterol (7α,25-OHC). The role of GPR183 in the central nervous system is not well characterized, and its role in pain is unknown. The profile of commercially available probes for GPR183 limits their use as pharmacological tools to dissect the roles of this receptor in pathophysiological settings. Using in silico modeling, we have screened a library of 5 million compounds to identify several novel small-molecule antagonists of GPR183 with nanomolar potency. These compounds are able to antagonize 7α,25-OHC-induced calcium mobilization in vitro with IC50 values below 50 nM. In vivo intrathecal injections of these antagonists during peak pain after CCI surgery reversed allodynia in male and female mice. Acute intrathecal injection of the GPR183 ligand 7α,25-OHC in naïve mice induced dose-dependent allodynia. Importantly, this effect was blocked using our novel GPR183 antagonists, suggesting spinal GPR183 activation as pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify this receptor as a potential target for therapeutic intervention. SIGNIFICANCE STATEMENT: We have identified several novel GPR183 antagonists with nanomolar potency. Using these antagonists, we have demonstrated that GPR183 signaling in the spinal cord is pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify it as a potential target for therapeutic intervention.


Assuntos
Neuralgia/metabolismo , Oxisteróis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Medula Espinal/metabolismo , Animais , Feminino , Células HL-60 , Humanos , Masculino , Camundongos , Neuralgia/tratamento farmacológico , Neuralgia/patologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais , Medula Espinal/patologia
5.
J Pharmacol Exp Ther ; 374(2): 331-341, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32434943

RESUMO

Treating chronic pain by using opioids, such as morphine, is hampered by the development of opioid-induced hyperalgesia (OIH; increased pain sensitivity), antinociceptive tolerance, and withdrawal, which can contribute to dependence and abuse. In the central nervous system, the purine nucleoside adenosine has been implicated in beneficial and detrimental actions of morphine, but the extent of their interaction remains poorly understood. Here, we demonstrate that morphine-induced OIH and antinociceptive tolerance in rats is associated with a twofold increase in adenosine kinase (ADK) expression in the dorsal horn of the spinal cord. Blocking ADK activity in the spinal cord provided greater than 90% attenuation of OIH and antinociceptive tolerance through A3 adenosine receptor (A3AR) signaling. Supplementing adenosine signaling with selective A3AR agonists blocked OIH and antinociceptive tolerance in rodents of both sexes. Engagement of A3AR in the spinal cord with an ADK inhibitor or A3AR agonist was associated with reduced dorsal horn of the spinal cord expression of the NOD-like receptor pyrin domain-containing 3 (60%-75%), cleaved caspase 1 (40%-60%), interleukin (IL)-1ß (76%-80%), and tumor necrosis factor (50%-60%). In contrast, the neuroinhibitory and anti-inflammatory cytokine IL-10 increased twofold. In mice, A3AR agonists prevented the development of tolerance in a model of neuropathic pain and reduced naloxone-dependent withdrawal behaviors by greater than 50%. These findings suggest A3AR-dependent adenosine signaling is compromised during sustained morphine to allow the development of morphine-induced adverse effects. These findings raise the intriguing possibility that A3AR agonists may be useful adjunct to opioids to manage their unwanted effects. SIGNIFICANCE STATEMENT: The development of hyperalgesia and antinociceptive tolerance during prolonged opioid use are noteworthy opioid-induced adverse effects that reduce opioid efficacy for treating chronic pain and increase the risk of dependence and abuse. We report that in rodents, these adverse effects are due to reduced adenosine signaling at the A3AR, resulting in NOD-like receptor pyrin domain-containing 3-interleukin-1ß neuroinflammation in spinal cord. These effects are attenuated by A3AR agonists, suggesting that A3AR may be a target for therapeutic intervention with selective A3AR agonist as opioid adjuncts.


Assuntos
Analgésicos/efeitos adversos , Tolerância a Medicamentos , Hiperalgesia/induzido quimicamente , Morfina/efeitos adversos , Receptor A3 de Adenosina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/etiologia , Adenosina/metabolismo , Animais , Feminino , Hiperalgesia/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/biossíntese , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
J Clin Invest ; 130(5): 2587-2592, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31999650

RESUMO

Treating neuropathic pain is challenging and novel non-opioid-based medicines are needed. Using unbiased receptomics, transcriptomic analyses, immunofluorescence, and in situ hybridization, we found that the expression of the orphan GPCR Gpr160 and GPR160 increased in the rodent dorsal horn of the spinal cord following traumatic nerve injury. Genetic and immunopharmacological approaches demonstrated that GPR160 inhibition in the spinal cord prevented and reversed neuropathic pain in male and female rodents without altering normal pain response. GPR160 inhibition in the spinal cord attenuated sensory processing in the thalamus, a key relay in the sensory discriminative pathways of pain. We also identified cocaine- and amphetamine-regulated transcript peptide (CARTp) as a GPR160 ligand. Inhibiting endogenous CARTp signaling in spinal cord attenuated neuropathic pain, whereas exogenous intrathecal CARTp evoked painful hypersensitivity through GPR160-dependent ERK and cAMP response element-binding protein (CREB). Our findings de-orphanize GPR160, identify it as a determinant of neuropathic pain and potential therapeutic target, and provide insights into its signaling pathways. CARTp is involved in many diseases including depression and reward and addiction; de-orphanization of GPR160 is a major step forward understanding the role of CARTp signaling in health and disease.


Assuntos
Neuralgia/etiologia , Neuralgia/fisiopatologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Linhagem Celular , Feminino , Humanos , Ligantes , Masculino , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/genética , Células PC12 , RNA Interferente Pequeno/genética , Ratos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Nervo Isquiático/lesões , Nervo Isquiático/fisiopatologia , Transdução de Sinais , Medula Espinal/metabolismo , Regulação para Cima
7.
Pain ; 161(1): 177-184, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31490328

RESUMO

Chemotherapy-induced neuropathic pain (CINP) in both sexes compromises many current chemotherapeutics and lacks an FDA-approved therapy. We recently identified the sphingosine-1-phosphate receptor subtype 1 (S1PR1) and A3 adenosine receptor subtype (A3AR) as novel targets for therapeutic intervention. Our work in male rodents using paclitaxel, oxaliplatin, and bortezomib showed robust inhibition of CINP with either S1PR1 antagonists or A3AR agonists. The S1PR1 functional antagonist FTY720 (Gilenya) is FDA-approved for treating multiple sclerosis, and selective A3AR agonists are in advanced clinical trials for cancer and inflammatory disorders, underscoring the need for their expedited trials in patients with CINP as chemotherapy adjuncts. Our findings reveal that S1PR1 antagonists and A3AR agonists mitigate paclitaxel and oxaliplatin CINP in female and male rodents, but failed to block or reverse bortezomib-induced neuropathic pain (BINP) in females. Although numerous mechanisms likely underlie these differences, we focused on receptor levels. We found that BINP in male rats, but not in female rats, was associated with increased expression of A3AR in the spinal cord dorsal horn, whereas S1PR1 levels were similar in both sexes. Thus, alternative mechanisms beyond receptor expression may account for sex differences in response to S1PR1 antagonists. Morphine and duloxetine, both clinical analgesics, reversed BINP in female mice, demonstrating that the lack of response is specific to S1PR1 and A3AR agents. Our findings suggest that A3AR- and S1PR1-based therapies are not viable approaches in preventing and treating BINP in females and should inform future clinical trials of these drugs as adjuncts to chemotherapy.


Assuntos
Antineoplásicos/efeitos adversos , Bortezomib/efeitos adversos , Neuralgia/tratamento farmacológico , Receptor A3 de Adenosina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Antagonistas do Receptor A3 de Adenosina/administração & dosagem , Antagonistas do Receptor A3 de Adenosina/uso terapêutico , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/uso terapêutico , Animais , Cloridrato de Duloxetina/administração & dosagem , Cloridrato de Duloxetina/uso terapêutico , Feminino , Cloridrato de Fingolimode/administração & dosagem , Cloridrato de Fingolimode/uso terapêutico , Masculino , Morfina/administração & dosagem , Morfina/uso terapêutico , Neuralgia/induzido quimicamente , Oxaliplatina/efeitos adversos , Paclitaxel/efeitos adversos , Ratos , Fatores Sexuais , Moduladores do Receptor de Esfingosina 1 Fosfato/administração & dosagem , Moduladores do Receptor de Esfingosina 1 Fosfato/uso terapêutico , Corno Dorsal da Medula Espinal/efeitos dos fármacos
8.
J Exp Med ; 215(5): 1301-1313, 2018 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-29703731

RESUMO

The development of chemotherapy-induced painful peripheral neuropathy is a major dose-limiting side effect of many chemotherapeutics, including bortezomib, but the mechanisms remain poorly understood. We now report that bortezomib causes the dysregulation of de novo sphingolipid metabolism in the spinal cord dorsal horn to increase the levels of sphingosine-1-phosphate (S1P) receptor 1 (S1PR1) ligands, S1P and dihydro-S1P. Accordingly, genetic and pharmacological disruption of S1PR1 with multiple S1PR1 antagonists, including FTY720, blocked and reversed neuropathic pain. Mice with astrocyte-specific alterations of S1pr1 did not develop neuropathic pain and lost their ability to respond to S1PR1 inhibition, strongly implicating astrocytes as a primary cellular substrate for S1PR1 activity. At the molecular level, S1PR1 engaged astrocyte-driven neuroinflammation and altered glutamatergic homeostasis, processes blocked by S1PR1 antagonism. Our findings establish S1PR1 as a target for therapeutic intervention and provide insight into cellular and molecular pathways. As FTY720 also shows promising anticancer potential and is FDA approved, rapid clinical translation of our findings is anticipated.


Assuntos
Bortezomib/efeitos adversos , Neuralgia/induzido quimicamente , Neuralgia/metabolismo , Esfingolipídeos/metabolismo , Administração Oral , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Ceramidas/biossíntese , Cloridrato de Fingolimode/administração & dosagem , Cloridrato de Fingolimode/farmacologia , Glutamatos/metabolismo , Masculino , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia
9.
Pain ; 159(6): 1025-1034, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29419652

RESUMO

Development of chemotherapy-induced neuropathic pain (CINP) compromises the use of chemotherapy and greatly impacts thousands of lives. Unfortunately, there are no Food and Drug Administration-approved drugs to prevent or treat CINP. Neuropathological changes within CNS, including neuroinflammation and increased neuronal excitability, are driven by alterations in neuro-glia communication; but, the molecular signaling pathways remain largely unexplored. Adenosine is a potent neuroprotective purine nucleoside released to counteract the consequences of these neuropathological changes. Adenosine signaling at its adenosine receptors (ARs) is dictated by adenosine kinase (ADK) in astrocytes, which provides a cellular sink for the removal of extracellular adenosine. We now demonstrate that chemotherapy (oxaliplatin) in rodents caused ADK overexpression in reactive astrocytes and reduced adenosine signaling at the A3AR subtype (A3AR) within the spinal cord. Dysregulation of ADK and A3AR signaling was associated with increased proinflammatory and neuroexcitatory interleukin-1ß expression and activation of nucleotide-binding oligomerization domain-like receptor protein 3 inflammasome, but not putative oxaliplatin-associated GSK3ß transcriptional regulation. Intrathecal administration of the highly selective A3AR agonist MRS5698 attenuated IL-1ß production and increased the expression of potent anti-inflammatory and neuroprotective IL-10. The effects of MRS5698 were blocked by attenuating IL-10 signaling in rats with intrathecal neutralizing IL-10 antibody and in IL-10 knockout mice. These findings provide new molecular insights implicating astrocyte-based ADK-adenosine axis and nucleotide-binding oligomerization domain-like receptor protein 3 in the development of CINP and IL-10 in the mechanism of action of A3AR agonists. These findings strengthen the pharmacological rationale for clinical evaluation of A3AR agonists already in advanced clinical trials as anticancer agents as an adjunct to chemotherapy.


Assuntos
Adenosina Quinase/metabolismo , Antineoplásicos/toxicidade , Astrócitos/metabolismo , Neuralgia/induzido quimicamente , Neuralgia/fisiopatologia , Oxaliplatina/toxicidade , Medula Espinal/enzimologia , Animais , Astrócitos/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Hiperalgesia/fisiopatologia , Interleucina-10/deficiência , Interleucina-10/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
10.
Pain ; 158(9): 1733-1742, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28570482

RESUMO

Metastatic bone pain is the single most common form of cancer pain and persists as a result of peripheral and central inflammatory, as well as neuropathic mechanisms. Here, we provide the first characterization of sphingolipid metabolism alterations in the spinal cord occurring during cancer-induced bone pain (CIBP). Following femoral arthrotomy and syngenic tumor implantation in mice, ceramides decreased with corresponding increases in sphingosine and the bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P). Intriguingly, de novo sphingolipid biosynthesis was increased as shown by the elevations of dihydro-ceramides and dihydro-S1P. We next identified the S1P receptor subtype 1 (S1PR1) as a novel target for therapeutic intervention. Intrathecal or systemic administration of the competitive and functional S1PR1 antagonists, TASP0277308 and FTY720/Fingolimod, respectively, attenuated cancer-induced spontaneous flinching and guarding. Inhibiting CIBP by systemic delivery of FTY720 did not result in antinociceptive tolerance over 7 days. FTY720 administration enhanced IL-10 in the lumbar ipsilateral spinal cord of CIBP animals and intrathecal injection of an IL-10 neutralizing antibody mitigated the ability of systemic FTY720 to reverse CIBP. FTY720 treatment was not associated with alterations in bone metabolism in vivo. Studies here identify a novel mechanism to inhibit bone cancer pain by blocking the actions of the bioactive metabolites S1P and dihydro-S1P in lumbar spinal cord induced by bone cancer and support potential fast-track clinical application of the FDA-approved drug, FTY720, as a therapeutic avenue for CIBP.


Assuntos
Dor do Câncer/etiologia , Dor do Câncer/metabolismo , Inflamação Neurogênica/etiologia , Inflamação Neurogênica/metabolismo , Pró-Proteína Convertases/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Serina Endopeptidases/metabolismo , Animais , Neoplasias da Mama/complicações , Neoplasias da Mama/patologia , Dor do Câncer/tratamento farmacológico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Inflamação Neurogênica/tratamento farmacológico , Pró-Proteína Convertases/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Sulfonas/farmacologia , Sulfonas/uso terapêutico , Triazóis/farmacologia , Triazóis/uso terapêutico
11.
Innate Immun ; 18(3): 373-89, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21862597

RESUMO

Yersinia pestis, a Gram-negative bacillus causing plague and Centers for Disease Control and Prevention (CDC) classified Category A pathogen, has high potential as a bioweapon. Lipopolysaccharide, a virulence factor for Y. pestis, binds to and activates A(1) adenosine receptor (AR)s and, in animals, A(1)AR antagonists block induced acute lung injury (ALI) and increase survival following cecal ligation and perforation. In this study, rats were infected intratracheally with viable Y. pestis [CO99 (pCD1( + )/Δpgm) 1 × 10( 8 ) CFU/animal] and treated daily for 3 d with ciprofloxacin (cipro), the A(1)AR antagonist L-97-1, or cipro plus L-97-1 starting at 0, 6, 24, 48, or 72 h post-Y. pestis. At 72 h post-Y. pestis, cipro plus L-97-1 significantly improved 6-d survival to 60-70% vs 28% for cipro plus H(2)O and 33% for untreated Y. pestis controls (P = 0.02, logrank test). Lung edema, hemorrhage and leukocyte infiltration index (LII) were evaluated histologically to produce ALI scores. Cipro plus L-97-1 significantly reduced lung edema, as well as aggregate lung injury scores vs controls or cipro plus H(2)O, and LII vs controls (P < 0.05, Student's unpaired t test). These results support efficacy for L-97-1 as a post-exposure medical countermeasure, adjunctive therapy to antibiotics for Y. pestis.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Pulmão/efeitos dos fármacos , Peste/tratamento farmacológico , Purinas/administração & dosagem , Yersinia pestis/imunologia , Lesão Pulmonar Aguda/etiologia , Animais , Ciprofloxacina/administração & dosagem , Ciprofloxacina/efeitos adversos , Modelos Animais de Doenças , Quimioterapia Combinada , Humanos , Lipopolissacarídeos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Peste/complicações , Peste/imunologia , Ligação Proteica/efeitos dos fármacos , Purinas/efeitos adversos , Ratos , Receptor A1 de Adenosina/metabolismo , Fatores de Virulência , Yersinia pestis/efeitos dos fármacos , Yersinia pestis/patogenicidade
12.
Respir Physiol Neurobiol ; 172(1-2): 63-71, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20470909

RESUMO

Reductions in alveolar oxygenation during lung hypoxia/reoxygenation (H/R) injury are common after gram-negative endotoxemia. However, the effects of H/R on endotoxin-stimulated cytokine production by alveolar macrophages are unclear and may depend upon thresholds for hypoxic oxyradical generation in situ. Here TNF-alpha and IL-1beta production were determined in rat alveolar macrophages stimulated with Escherichia coli lipopolysaccharide (LPS, serotype O55:B5) while exposed to either normoxia for up to 24h, to brief normocarbic hypoxia (1.5h at an atmospheric PO(2)=10+/-2mm Hg), or to combined H/R. LPS-induced TNF-alpha and IL-1beta were reduced at the peak of hypoxia and by reoxygenation in LPS+H/R cells (P<0.01) compared with normoxic controls despite no changes in reduced glutathione (GSH) or in PGE2 production. Both TNF-alpha mRNA and NF-kappaB activation were reduced by hypoxia that suppressed superoxide anion generation. Thus, dynamic reductions in the ambient PO(2) of alveolar macrophages that do not deplete GSH suppress LPS-induced TNF-alpha expression, IL-1beta production, and NF-kappaB activation even as oxyradical production is decreased.


Assuntos
Hipóxia Celular/fisiologia , Citocinas/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , NF-kappa B/metabolismo , Análise de Variância , Animais , Linhagem Celular Transformada , Citocinas/genética , Dinoprostona/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , L-Lactato Desidrogenase/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo
13.
Crit Care Med ; 38(7): 1574-83, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20400901

RESUMO

OBJECTIVE: Pneumonic plague resulting from Yersinia pestis induces swiftly lethal sepsis and is a major concern as a weapon of bioterrorism. However, the role of specific plasmid-encoded vs. chromosomal Y. pestis virulence factors in the pathogenesis of acute lung injury, shock, and nonpulmonary dysfunction is unclear. We hypothesized that the pathophysiology of pneumonic plague resulting from expression of proteins encoded by the thermally regulated pCD1 plasmid differs from cardiopulmonary and inflammatory changes attributable to the chromosomal pgm gene locus. DESIGN: Prospective, experimental study. SETTING: Research laboratory at a university medical center. SUBJECTS: Conscious, chronically catheterized male Sprague-Dawley rats (total n=104). INTERVENTIONS: Rats were intratracheally infected with 109 colony-forming units of Y. pestis attenuated strains CO99 (pCD1+/DeltaApgm) or KIM6+ (pCD1-/pgm+) and evaluated over 6 days. Serial evaluations of vital signs, cardiorespiratory parameters, blood cultures, inflammatory biomarkers, and organ function were obtained, as well as organ histopathology and cytokine production. MEASUREMENTS AND MAIN RESULTS: Despite equivalent endotoxin contents between the inocula, CO99-infected animals had a median survival of 3 days with greater nonpulmonary organ injury, microbial growth, serum alanine aminotransferase, and liver microvascular permeability vs. KIM6+-infected animals (p<.05). Parallel differences occurred in serum tumor necrosis factor-alpha levels. Notably, 75% of CO99 rats developed fatal hypotension after developing nonpulmonary organ damage. CONCLUSION: These results suggest that progression to lethal sepsis with augmented liver injury during plague pneumonia requires factors encoded by the pCD1 plasmid but not chromosomal proteins present within the pgm gene locus.


Assuntos
Proteínas de Bactérias/fisiologia , Insuficiência de Múltiplos Órgãos/fisiopatologia , Peste/fisiopatologia , Choque Séptico/fisiopatologia , Fatores de Virulência/fisiologia , Yersinia pestis/fisiologia , Alanina Transaminase/sangue , Animais , Proteínas de Bactérias/genética , Permeabilidade Capilar , Citocinas/sangue , Ensaio de Imunoadsorção Enzimática , Genes Bacterianos , Loci Gênicos , Hemodinâmica , Fígado/irrigação sanguínea , Masculino , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/microbiologia , Peste/complicações , Peste/microbiologia , Plasmídeos , Ratos , Ratos Sprague-Dawley , Choque Séptico/etiologia , Choque Séptico/microbiologia , Fatores de Virulência/genética , Yersinia pestis/genética
14.
Shock ; 23(2): 186-93, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15665736

RESUMO

Gram-negative bacterial infection predisposes to the development of shock and acute lung injury with multiple organ dysfunction in the critically ill. Although overexpression of proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha) and interleukin (IL)-1beta, IL-6, IL-8, and other mediators is causally implicated in the pathogenesis of shock and lung injury, the underlying mechanisms following cellular exposure to gram-negative endotoxin remain unclear. De novo generation of reactive oxygen species (ROS) by monocytes/macrophages in particular has been proposed as a pivotal regulatory mechanism by which enhanced transactivation of redox-sensitive genes culminates in augmented cytokine expression within the lower respiratory tract. Here we sought to characterize the mechanism of action of a synthetic, nonpeptide, low-molecular-weight, Mn-containing superoxide dismutase mimetic (SODm), M40403, in modulating E. coli lipopolysaccharide serotype 0111:B4 (LPS)-induced cytokine production by cultured rat alveolar macrophages. Intracellular superoxide (O2) ion generation was measured using hydroethidine (HE) dye, and the dose-dependent effects of M40403 on TNF-alpha and IL-6 biosynthesis by ELISAs. Upstream redox-sensitive signaling events involving the pleiotropic transcription factor NF-kappaB were determined in nuclear extracts by electrophoretic mobility shift assays (EMSAs) and p65 subunit Western blot. The levels of the cytosolic inhibitory protein IkappaB-alpha were also assessed by Western analysis. We found that M40403 potently suppressed the production of superoxide, TNF-alpha, and IL-6 in LPS-stimulated alveolar macrophages, suggesting a key role for superoxide in endotoxin-induced cytokine production in the distal air spaces. In addition, M40403 decreased E. coli LPS-induced activation of NF-kappaB, and this effect was associated with modest suppression of cytoplasmic IkappaB-alpha degradation. Together, these results suggest that removal of superoxide by M40403 inhibits endotoxin-induced production of TNF-alpha and IL-6 in alveolar macrophages by a mechanism involving suppression of redox-sensitive NF-kappaB transactivation or signaling.


Assuntos
Citocinas/metabolismo , Endotoxinas/metabolismo , NF-kappa B/metabolismo , Superóxidos/metabolismo , Animais , Western Blotting , Núcleo Celular/metabolismo , Corantes/farmacologia , Citocinas/biossíntese , Citoplasma/metabolismo , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Escherichia coli/metabolismo , Proteínas I-kappa B/metabolismo , Immunoblotting , Inflamação , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Macrófagos Alveolares/metabolismo , Manganês/metabolismo , Modelos Biológicos , Inibidor de NF-kappaB alfa , Compostos Organometálicos/farmacologia , Oxirredução , Oxigênio/metabolismo , Fenantridinas/farmacologia , Ratos , Espécies Reativas de Oxigênio , Transdução de Sinais , Superóxido Dismutase/metabolismo , Fatores de Tempo , Fator de Transcrição RelA , Fator de Necrose Tumoral alfa/metabolismo
15.
Am J Physiol Regul Integr Comp Physiol ; 287(2): R437-45, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15059791

RESUMO

Transactivation of the DNA-binding proteins nuclear factor-kappa B (NF-kappa B) and activator protein (AP)-1 by de novo oxyradical generation is a stereotypic redox-sensitive process during hypoxic stress of the liver. Systemic trauma is associated with splanchnic hypoxia-reoxygenation (H/R) followed by intraportal gram-negative bacteremia, which collectively have been implicated in posttraumatic liver dysfunction and multiple organ damage. We hypothesized that hypoxic stress of the liver before stimulation by Escherichia coli serotype O55:B5 (EC) amplifies oxyradical-mediated transactivation of NF-kappa B and AP-1 as well as cytokine production compared with noninfectious H/R or gram-negative sepsis without prior hypoxia. Livers from Sprague-Dawley rats underwent perfusion for 180 min with or without 0.5 h of hypoxia (perfusate PO(2), 40 +/- 5 mmHg) followed by reoxygenation and infection with 10(9) EC or 0.9% NaCl infusion. In H/R + EC livers, nuclear translocation of NF-kappa B and AP-1 was unexpectedly reduced in gel shift assays vs. normoxic EC controls, as were perfusate TNF-alpha and IL-1 beta levels. Preceding hypoxic stress paradoxically increased postbacteremic reduced-to-oxidized glutathione ratios plus nuclear localization of I kappa B alpha and phospho-I kappa B alpha, but not JunB/FosB profiles. Notably, xanthine oxidase inhibition increased transactivation as well as cytokine production in H/R + EC livers. Thus brief hypoxic stress of the liver before intraportal gram-negative bacteremia potently suppresses activation of canonical redox-sensitive transcription factors and production of inflammatory cytokines by mechanisms including xanthine oxidase-induced oxyradicals functioning in an anti-inflammatory signaling role. These results suggest a novel multifunctionality of oxyradicals in decoupling hepatic transcriptional activity and cytokine biosynthesis early in the posttraumatic milieu.


Assuntos
Infecções por Escherichia coli/metabolismo , Hipóxia/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição AP-1/metabolismo , Animais , Núcleo Celular/metabolismo , Citocinas/metabolismo , Citoplasma/metabolismo , Infecções por Escherichia coli/fisiopatologia , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Hipóxia/fisiopatologia , Proteínas I-kappa B/metabolismo , Fígado/metabolismo , Masculino , Inibidor de NF-kappaB alfa , NF-kappa B/genética , Oxirredução , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição AP-1/genética , Transcrição Gênica/fisiologia , Xantina Oxidase/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA