Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 21(12): 1798-1809, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36190955

ABSTRACT

Multiple myeloma is a plasma cell malignancy that is still largely incurable, despite considerable progress in recent years. NF-κB is a well-established therapeutic target in multiple myeloma, but none of the currently available treatment options offer direct, specific pharmacologic targeting of NF-κB transcriptional activity. Thus, we designed a novel direct NF-κB inhibitor (IT848) as a drug candidate with strong potential for clinical translation and conducted comprehensive in vitro and in vivo mechanistic studies in multiple myeloma cell lines, primary multiple myeloma cells, xenograft models, and immunocompetent mouse models of multiple myeloma. Here, we show that IT848 inhibits NF-κB activity through inhibition of DNA binding of all five NF-κB subunits. IT848 treatment of multiple myeloma cell lines and patient samples inhibited proliferation and induced caspase-dependent and independent apoptosis. In addition to direct NF-κB inhibitory effects, IT848 treatment altered the redox homeostasis of multiple myeloma cells through depletion of the reduced glutathione pool, selectively inducing oxidative stress in multiple myeloma but not in healthy cells. Multiple myeloma xenograft studies confirmed the efficacy of IT848 as single agent and in combination with bortezomib. Furthermore, IT848 significantly improved survival when combined with programmed death protein 1 inhibition, and correlative immune studies revealed that this clinical benefit was associated with suppression of regulatory T-cell infiltration of the bone marrow microenvironment. In conclusion, IT848 is a potent direct NF-κB inhibitor and inducer of oxidative stress specifically in tumor cells, displaying significant activity against multiple myeloma cells in vitro and in vivo, both as monotherapy as well as in combination with bortezomib or immune checkpoint blockade.


Subject(s)
Multiple Myeloma , Mice , Animals , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Bortezomib/pharmacology , Bortezomib/therapeutic use , Tumor Microenvironment , Apoptosis , I-kappa B Proteins/metabolism , Oxidation-Reduction , DNA/metabolism , Cell Line, Tumor
3.
Transplant Cell Ther ; 27(5): 391-403, 2021 05.
Article in English | MEDLINE | ID: mdl-33965177

ABSTRACT

Autologous hematopoietic stem cell transplantation (ASCT) is a standard-of-care treatment for many hematologic malignancies. Progression of disease after ASCT is the primary cause of treatment failure. In this Phase Ib trial, we studied the safety and clinical effect of combined checkpoint inhibition therapy (CPIT) with ipilimumab and nivolumab as a consolidation strategy after ASCT for patients with high-risk diffuse large B cell lymphoma (DLBCL), mature T cell lymphoma (TCL), and multiple myeloma (MM). Starting at 14 to 28 days after ASCT, patients received ipilimumab (1 mg/kg i.v. on day 1 of weeks 1, 4, 7, 10, 16, and 22) and nivolumab (3 mg/kg i.v. on day 1 of weeks 1, 4, 7, 10, 12, 14, 16, 18, 20, 22, 24, and 26). Patients received a median of 5 doses of ipilimumab and 8 doses of nivolumab. Thirty-five patients were included in the intent-to-treat population. Ninety-four percent of the patients experienced immune-related adverse events (irAEs) of any grade. Ninety-seven percent of irAEs resolved spontaneously or after holding study drugs and instituting high-dose corticosteroid therapy. Progression-free and overall survival at 18 months post-ASCT for each disease cohort were 85.7% and 100% for primary refractory DLBCL, 28.6% and 57.1% for relapsed DLBCL, not evaluable and 80% for frontline TCL, 25% and 75% for relapsed TCL, 57.1% and 87% for high-risk transplant-naïve MM, and 40% and 100% for MM relapsed within 3 years of first ASCT. We conclude that combined CPIT appears to be tolerable as a consolidation strategy after ASCT and in addition to the potential clinical efficacy observed in some subsets of disease, T cell receptor repertoire, T regulatory cell phenotype, and gut microbiota profiles provide a biologic rationale warranting further study of this approach.


Subject(s)
Hematopoietic Stem Cell Transplantation , Consolidation Chemotherapy , Humans , Ipilimumab/adverse effects , Neoplasm Recurrence, Local , Nivolumab , Transplantation, Autologous
4.
JCI Insight ; 6(6)2021 03 22.
Article in English | MEDLINE | ID: mdl-33571168

ABSTRACT

Here, we report on a phase IIa study to determine the intubation rate, survival, viral clearance, and development of endogenous Abs in patients with COVID-19 pneumonia treated with convalescent plasma (CCP) containing high levels of neutralizing anti-SARS-CoV-2 Abs. Radiographic and laboratory evaluation confirmed all 51 treated patients had COVID-19 pneumonia. Fresh or frozen CCP from donors with high titers of neutralizing Abs was administered. The nonmechanically ventilated patients (n = 36) had an intubation rate of 13.9% and a 30-day survival rate of 88.9%, and the overall survival rate for a comparative group based on network data was 72.5% (1625/2241). Patients had negative nasopharyngeal swab rates of 43.8% and 73.0% on days 10 and 30, respectively. Patients mechanically ventilated had a day-30 mortality rate of 46.7%; the mortality rate for a comparative group based on network data was 71.0% (369/520). All evaluable patients were found to have neutralizing Abs on day 3 (n = 47), and all but 1 patient had Abs on days 30 and 60. The only adverse event was a mild rash. In this study on patients with COVID-19 disease, we show therapeutic use of CCP was safe and conferred transfer of Abs, while preserving endogenous immune response.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , COVID-19/therapy , Immunoglobulin G/therapeutic use , Plasma , SARS-CoV-2/immunology , Severity of Illness Index , Aged , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/immunology , COVID-19/mortality , COVID-19/virology , Convalescence , Female , Humans , Immunization, Passive , Immunocompromised Host , Immunoglobulin G/blood , Male , Middle Aged , Pneumonia , Respiration, Artificial , COVID-19 Serotherapy
5.
Genome Biol ; 21(1): 153, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32594908

ABSTRACT

BACKGROUND: Mapping of allele-specific DNA methylation (ASM) can be a post-GWAS strategy for localizing regulatory sequence polymorphisms (rSNPs). The advantages of this approach, and the mechanisms underlying ASM in normal and neoplastic cells, remain to be clarified. RESULTS: We perform whole genome methyl-seq on diverse normal cells and tissues and three cancer types. After excluding imprinting, the data pinpoint 15,112 high-confidence ASM differentially methylated regions, of which 1838 contain SNPs in strong linkage disequilibrium or coinciding with GWAS peaks. ASM frequencies are increased in cancers versus matched normal tissues, due to widespread allele-specific hypomethylation and focal allele-specific hypermethylation in poised chromatin. Cancer cells show increased allele switching at ASM loci, but disruptive SNPs in specific classes of CTCF and transcription factor binding motifs are similarly correlated with ASM in cancer and non-cancer. Rare somatic mutations affecting these same motif classes track with de novo ASM. Allele-specific transcription factor binding from ChIP-seq is enriched among ASM loci, but most ASM differentially methylated regions lack such annotations, and some are found in otherwise uninformative "chromatin deserts." CONCLUSIONS: ASM is increased in cancers but occurs by a shared mechanism involving disruptive SNPs in CTCF and transcription factor binding sites in both normal and neoplastic cells. Dense ASM mapping in normal plus cancer samples reveals candidate rSNPs that are difficult to find by other approaches. Together with GWAS data, these rSNPs can nominate specific transcriptional pathways in susceptibility to autoimmune, cardiometabolic, neuropsychiatric, and neoplastic diseases.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA Methylation , Neoplasms/metabolism , Transcription Factors/metabolism , Alleles , CpG Islands , Genomic Imprinting , Humans , Linkage Disequilibrium , Neoplasms/genetics , Polymorphism, Single Nucleotide , Whole Genome Sequencing
6.
Biol Blood Marrow Transplant ; 21(6): 1000-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25459643

ABSTRACT

Allogeneic blood and marrow transplantation (allo-BMT) is an effective immunotherapeutic treatment that can provide partial or complete remission for patients with hematological malignancies. Mature donor T cells in the donor inoculum play a central role in mediating graft-versus-tumor (GVT) responses by destroying residual tumor cells that persist after conditioning regimens. Alloreactivity towards minor histocompatibility antigens (miHA), which are varied tissue-related self-peptides presented in the context of major histocompatibility complex (MHC) molecules on recipient cells, some of which may be shared on tumor cells, is a dominant factor for the development of GVT. Potentially, GVT can also be directed to tumor-associated antigens or tumor-specific antigens that are more specific to the tumor cells themselves. The full exploitation of allo-BMT, however, is greatly limited by the development of graft-versus-host disease (GVHD), which is mediated by the donor T cell response against the miHA expressed in the recipient's cells of the intestine, skin, and liver. Because of the significance of GVT and GVHD responses in determining the clinical outcome of patients, miHA and tumor antigens have been intensively studied, and one active immunotherapeutic approach to separate these two responses has been cancer vaccination after allo-BMT. The combination of these two strategies has an advantage over vaccination of the patient without allo-BMT because his or her immune system has already been exposed and rendered unresponsive to the tumor antigens. The conditioning for allo-BMT eliminates the patient's existing immune system, including regulatory elements, and provides a more permissive environment for the newly developing donor immune compartment to selectively target the malignant cells. Utilizing recent technological advances, the identities of many human miHA and tumor antigenic peptides have been defined and are currently being evaluated in clinical and basic immunological studies for their ability to produce effective T cell responses. The first step towards this goal is the identification of targetable tumor antigens. In this review, we will highlight some of the technologies currently used to identify tumor antigens and anti-tumor T cell clones in hematological malignancies.


Subject(s)
Antigens, Neoplasm/analysis , Bone Marrow Transplantation , Graft vs Tumor Effect , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , T-Lymphocytes/immunology , Adoptive Transfer , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Biomarkers/analysis , Cancer Vaccines/administration & dosage , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , T-Lymphocytes/transplantation , Transplantation, Homologous , Treatment Outcome
7.
Shock ; 38(1): 107-14, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22575992

ABSTRACT

Interactions of toll-like receptors (TLRs) with nonmicrobial factors play a major role in the pathogenesis of early trauma-hemorrhagic shock (T/HS)-induced organ injury and inflammation. Thus, we tested the hypothesis that TLR4 mutant (TLR4 mut) mice would be more resistant to T/HS-induced gut injury and polymorphonuclear neutrophil (PMN) priming than their wild-type littermates and found that both were significantly reduced in the TLR4 mut mice. In addition, the in vivo and ex vivo PMN priming effect of T/HS intestinal lymph observed in the wild-type mice was abrogated in TLR4 mut mice as well the TRIF mut-deficient mice and partially attenuated in Myd88 mice, suggesting that TRIF activation played a more predominant role than MyD88 in T/HS lymph-induced PMN priming. Polymorphonuclear neutrophil depletion studies showed that T/HS lymph-induced acute lung injury was PMN dependent, because lung injury was totally abrogated in PMN-depleted animals. Because the lymph samples were sterile and devoid of endotoxin or bacterial DNA, we investigated whether the effects of T/HS lymph was related to endogenous nonmicrobial TLR4 ligands. High-mobility group box 1 protein 1, heat shock protein 70, heat shock protein 27, and hyaluronic acid all have been implicated in ischemia-reperfusion-induced tissue injury. None of these "danger" proteins appeared to be involved, because their levels were similar between the sham and shock lymph samples. In conclusion, TLR4 activation is important in T/HS-induced gut injury and in T/HS lymph-induced PMN priming and lung injury. However, the T/HS-associated effects of TLR4 on gut barrier dysfunction can be uncoupled from the T/HS lymph-associated effects of TLR4 on PMN priming.


Subject(s)
Intestinal Diseases/etiology , Neutrophil Activation/immunology , Shock, Hemorrhagic/complications , Toll-Like Receptor 4/immunology , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Animals , Intestinal Diseases/immunology , Intestinal Diseases/physiopathology , Intestinal Mucosa/metabolism , Ligands , Lymph/immunology , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Permeability , Rats , Respiratory Burst/immunology , Shock, Hemorrhagic/immunology , Shock, Hemorrhagic/physiopathology , Signal Transduction/physiology , Swine , Swine, Miniature , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Wounds and Injuries/complications
8.
PLoS One ; 6(8): e14829, 2011.
Article in English | MEDLINE | ID: mdl-21829592

ABSTRACT

BACKGROUND: Injurious non-microbial factors released from the stressed gut during shocked states contribute to the development of acute lung injury (ALI) and multiple organ dysfunction syndrome (MODS). Since Toll-like receptors (TLR) act as sensors of tissue injury as well as microbial invasion and TLR4 signaling occurs in both sepsis and noninfectious models of ischemia/reperfusion (I/R) injury, we hypothesized that factors in the intestinal mesenteric lymph after trauma hemorrhagic shock (T/HS) mediate gut-induced lung injury via TLR4 activation. METHODS/PRINCIPAL FINDINGS: The concept that factors in T/HS lymph exiting the gut recreates ALI is evidenced by our findings that the infusion of porcine lymph, collected from animals subjected to global T/HS injury, into naïve wildtype (WT) mice induced lung injury. Using C3H/HeJ mice that harbor a TLR4 mutation, we found that TLR4 activation was necessary for the development of T/HS porcine lymph-induced lung injury as determined by Evan's blue dye (EBD) lung permeability and myeloperoxidase (MPO) levels as well as the induction of the injurious pulmonary iNOS response. TRIF and Myd88 deficiency fully and partially attenuated T/HS lymph-induced increases in lung permeability respectively. Additional studies in TLR2 deficient mice showed that TLR2 activation was not involved in the pathology of T/HS lymph-induced lung injury. Lastly, the lymph samples were devoid of bacteria, endotoxin and bacterial DNA and passage of lymph through an endotoxin removal column did not abrogate the ability of T/HS lymph to cause lung injury in naïve mice. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that non-microbial factors in the intestinal mesenteric lymph after T/HS are capable of recreating T/HS-induced lung injury via TLR4 activation.


Subject(s)
Lung Injury/etiology , Lymph Nodes/metabolism , Shock, Hemorrhagic/complications , Toll-Like Receptor 4/metabolism , Wounds and Injuries/complications , Animals , Base Sequence , Blotting, Western , DNA Primers , Lung/enzymology , Male , Mice , Nitric Oxide Synthase Type II/metabolism , Polymerase Chain Reaction , Signal Transduction , Swine , Swine, Miniature
9.
Shock ; 36(2): 177-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21558984

ABSTRACT

Many models of trauma-hemorrhagic shock (T/HS) involve the reinfusion of anticoagulated shed blood. Our recent observation that the anticoagulant heparin induces increased mesenteric lymph lipase activity and consequent in vitro endothelial cell cytotoxicity prompted us to investigate the effect of heparin-induced lipase activity on organ injury in vivo as well as the effects of other anticoagulants on mesenteric lymph bioactivity in vitro and in vivo. To investigate this issue, rats subjected to trauma-hemorrhage had their shed blood anticoagulated with heparin, the synthetic anticoagulant arixtra (fondaparinux sodium), or citrate. Arixtra, in contrast to heparin, did not increase lymph lipase activity or result in high levels of endothelial cytotoxicity. Yet, the arixtra-treated rats subjected to T/HS still manifested lung injury, neutrophil priming, and red blood cell dysfunction, which was totally abrogated by lymph duct ligation. Furthermore, the injection of T/HS mesenteric lymph, but not sham-shock lymph, collected from the arixtra rats into control mice recreated the pattern of lung injury, polymorphonucleocyte (PMN) priming, and red blood cell dysfunction observed after actual shock. Consistent with these observations, citrate-anticoagulated rats subjected to T/HS developed lung injury, and the injection of mesenteric lymph from the citrate-anticoagulated T/HS rats into control mice also resulted in lung injury. Based on these results, several conclusions can be drawn. First, heparin-induced increased mesenteric lymph lipase activity is not responsible for the in vivo effects of T/HS mesenteric lymph. Second, heparin should be avoided as an anticoagulant when studying the biology or composition of mesenteric lymph because of its ability to cause increases in lymph lipase activity that increase the in vitro cytotoxicity of these lymph samples.


Subject(s)
Anticoagulants/pharmacology , Lymph/drug effects , Lymph/metabolism , Shock, Hemorrhagic/metabolism , Animals , Cells, Cultured , Citric Acid/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fondaparinux , Heparin/pharmacology , Lipase/metabolism , Male , Polysaccharides/pharmacology , Rats , Rats, Sprague-Dawley
10.
Am J Physiol Gastrointest Liver Physiol ; 300(5): G853-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21183660

ABSTRACT

Gut injury and loss of normal intestinal barrier function are key elements in the paradigm of gut-origin systemic inflammatory response syndrome, acute lung injury, and multiple organ dysfunction syndrome (MODS). As hypoxia-inducible factor (HIF-1) is a critical determinant of the physiological and pathophysiological response to hypoxia and ischemia, we asked whether HIF-1 plays a proximal role in the induction of gut injury and subsequent lung injury. Using partially HIF-1α-deficient mice in an isolated superior mesenteric artery occlusion (SMAO) intestinal ischemia reperfusion (I/R) injury model (45 min SMAO followed by 3 h of reperfusion), we showed a direct relationship between HIF-1 activation and intestinal I/R injury. Specifically, partial HIF-1α deficiency attenuated SMAO-induced increases in intestinal permeability, lipid peroxidation, mucosal caspase-3 activity, and IL-1ß mRNA levels. Furthermore, partial HIF-1α deficiency prevented the induction of ileal mucosal inducible nitric oxide synthase (iNOS) protein levels after SMAO and iNOS deficiency ameliorated SMAO-induced villus injury. Resistance to SMAO-induced gut injury was also associated with resistance to lung injury, as reflected by decreased levels of myeloperoxidase, IL-6 and IL-10 in the lungs of HIF-1α(+/-) mice. In contrast, a short duration of SMAO (15 min) followed by 3 h of reperfusion neither induced mucosal HIF-1α protein levels nor caused significant gut and lung injury in wild-type or HIF-1α(+/-) mice. This study indicates that intestinal HIF-1 activation is a proximal regulator of I/R-induced gut mucosal injury and gut-induced lung injury. However, the duration and severity of the gut I/R insult dictate whether HIF-1 plays a gut-protective or deleterious role.


Subject(s)
Hypoxia-Inducible Factor 1/physiology , Intestinal Diseases/pathology , Reperfusion Injury/pathology , Acute Lung Injury/pathology , Animals , Blotting, Western , Caspase 3/metabolism , Enzyme-Linked Immunosorbent Assay , Genotype , Hypoxia-Inducible Factor 1/genetics , Intestinal Mucosa/physiology , Intestines/blood supply , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Permeability , Peroxidase/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Am J Physiol Gastrointest Liver Physiol ; 299(4): G833-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20689059

ABSTRACT

Acute lung injury (ALI) and the development of the multiple organ dysfunction syndrome (MODS) are major causes of death in trauma patients. Gut inflammation and loss of gut barrier function as a consequence of splanchnic ischemia-reperfusion (I/R) have been implicated as the initial triggering events that contribute to the development of the systemic inflammatory response, ALI, and MODS. Since hypoxia-inducible factor (HIF-1) is a key regulator of the physiological and pathophysiological response to hypoxia, we asked whether HIF-1 plays a proximal role in the induction of gut injury and subsequent lung injury. Utilizing partially HIF-1α-deficient mice in a global trauma hemorrhagic shock (T/HS) model, we found that HIF-1 activation was necessary for the development of gut injury and that the prevention of gut injury was associated with an abrogation of lung injury. Specifically, in vivo studies demonstrated that partial HIF-1α deficiency ameliorated T/HS-induced increases in intestinal permeability, bacterial translocation, and caspase-3 activation. Lastly, partial HIF-1α deficiency reduced TNF-α, IL-1ß, cyclooxygenase-2, and inducible nitric oxide synthase levels in the ileal mucosa after T/HS whereas IL-1ß mRNA levels were reduced in the lung after T/HS. This study indicates that prolonged intestinal HIF-1 activation is a proximal regulator of I/R-induced gut mucosal injury and gut-induced lung injury. Consequently, these results provide unique information on the initiating events in trauma-hemorrhagic shock-induced ALI and MODS as well as potential therapeutic insights.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation/metabolism , Intestinal Diseases/metabolism , Intestines/injuries , Reperfusion Injury/metabolism , Animals , Apoptosis , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/physiology , Genotype , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/pathology , Lung/metabolism , Lung/pathology , Lung Injury/metabolism , Lung Injury/pathology , Mice , Permeability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reperfusion Injury/pathology , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/pathology
12.
PLoS One ; 5(2): e9421, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20195535

ABSTRACT

BACKGROUND: Acute lung injury (ALI) and the development of the multiple organ dysfunction syndrome (MODS) is a major cause of death in trauma patients. Earlier studies in trauma hemorrhagic shock (T/HS) have documented that splanchnic ischemia leading to gut inflammation and loss of barrier function is an initial triggering event that leads to gut-induced ARDS and MODS. Since sex hormones have been shown to modulate the response to T/HS and proestrous (PE) females are more resistant to T/HS-induced gut and distant organ injury, the goal of our study was to determine the contribution of estrogen receptor (ER)alpha and ERbeta in modulating the protective response of female rats to T/HS-induced gut and lung injury. METHODS/PRINCIPAL FINDINGS: The incidence of gut and lung injury was assessed in PE and ovariectomized (OVX) female rats subjected to T/HS or trauma sham shock (T/SS) as well as OVX rats that were administered estradiol (E2) or agonists for ERalpha or ERbeta immediately prior to resuscitation. Marked gut and lung injury was observed in OVX rats subjected to T/HS as compared to PE rats or E2-treated OVX rats subjected to T/HS. Both ERalpha and ERbeta agonists were equally effective in limiting T/HS-induced morphologic villous injury and bacterial translocation, whereas the ERbeta agonist was more effective than the ERalpha agonist in limiting T/HS-induced lung injury as determined by histology, Evan's blue lung permeability, bronchoalevolar fluid/plasma protein ratio and myeloperoxidase levels. Similarly, treatment with either E2 or the ERbeta agonist attenuated the induction of the intestinal iNOS response in OVX rats subjected to T/HS whereas the ERalpha agonist was only partially protective. CONCLUSIONS/SIGNIFICANCE: Our study demonstrates that estrogen attenuates T/HS-induced gut and lung injury and that its protective effects are mediated by the activation of ERalpha, ERbeta or both receptors.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Multiple Organ Failure/prevention & control , Animals , Enterocytes/drug effects , Enterocytes/enzymology , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Estrogens/pharmacology , Estrous Cycle , Female , Immunohistochemistry , Intestines/injuries , Lung Injury/etiology , Lung Injury/physiopathology , Lung Injury/prevention & control , Multiple Organ Failure/etiology , Multiple Organ Failure/physiopathology , Nitric Oxide Synthase Type II/metabolism , Ovariectomy , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/complications , Shock, Traumatic/complications
13.
J Trauma ; 68(1): 35-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20065755

ABSTRACT

BACKGROUND: Decreased red blood cell (RBC) deformability and activation of neutrophils (polymorphonuclear leukocytes [PMN]) after trauma-hemorrhagic shock (T/HS) have been implicated in the development of multiple organ dysfunction. Experimentally, female animals seemed to be protected from the effects of T/HS, at least in part, because of elevated estrogen levels. Thus, we examined the relative role of estrogen receptor (ER)-alpha and -beta in this protective response. METHODS: To accomplish this goal, RBC deformability and neutrophil respiratory burst activity were measured in several groups of hormonally intact or ovariectomized (OVX) female rats subjected to T/HS (laparotomy plus hemorrhage to an MAP of 30 mm Hg to 35 mm Hg for 90 minutes) or trauma-sham shock (T/SS) and 3 hours of reperfusion. These groups included rats receiving vehicle, estradiol, or either an ER-alpha agonist or an ER-beta agonist administered at the end of the shock period just before volume resuscitation. RESULTS: RBC deformability and neutrophil activation were similar among all the T/SS groups and were not different from that observed in the non-OVX female rats subjected to T/HS. In contrast, RBC deformability was reduced and neutrophil activation was increased in the OVX, T/HS female rats as compared with the T/SS groups or the non-OVX, T/HS rats. The administration of estrogen to the T/HS, OVX rats returned RBC and neutrophil function to normal. Both the ER-alpha and -beta agonist partially, but not completely, protected the OVX rats from T/HS-induced loss of RBC deformability, whereas only the ER-beta agonist prevented the increase in neutrophil activation. CONCLUSIONS: The protective effects of estrogen on T/HS-induced RBC deformability are mediated, at least in part, via activation of both ER-alpha and -beta, whereas ER-beta activation is involved in limiting T/HS-induced neutrophil activation.


Subject(s)
Erythrocyte Deformability , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Shock, Hemorrhagic/blood , Shock, Traumatic/blood , Animals , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Female , Neutrophil Activation/drug effects , Neutrophils/metabolism , Nitriles/pharmacology , Ovariectomy , Phenols , Propionates/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Respiratory Burst , Shock, Hemorrhagic/physiopathology , Shock, Traumatic/physiopathology
14.
Am J Physiol Lung Cell Mol Physiol ; 296(3): L404-17, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19118093

ABSTRACT

Intestinal ischemia after trauma-hemorrhagic shock (T/HS) results in gut barrier dysfunction and the production/release of biologically active and tissue injurious factors in the mesenteric lymph, which, in turn, causes acute lung injury and a systemic inflammatory state. Since T/HS-induced lung injury is associated with pulmonary endothelial and epithelial cell programmed cell death (PCD) and was abrogated by mesenteric lymph duct ligation, we sought to investigate the cellular pathways involved. Compared with trauma-sham shock (T/SS) rats, a significant increase in caspase-3 and M30 expression was detected in the pulmonary epithelial cells undergoing PCD, whereas apoptosis-inducing factor (AIF), but not caspase-3, was detected in endothelial cells undergoing PCD. This AIF-mediated pulmonary endothelial PCD response was validated in an in situ femoral vein assay where endothelial cells were found to express AIF but not caspase-3. To complement these studies, human umbilical vein endothelial cell (HUVEC), human lung microvascular endothelial cell (HLMEC), and human alveolar type II epithelial cell (A549) lines were used as in vitro models. T/HS lymph induced the nuclear translocation of AIF in HUVEC and HLMEC, and caspase inhibition in these cells did not afford any cytoprotection. For proof of principle, AIF silencing in HUVEC reversed the cytotoxic effects of T/HS on cell viability and DNA fragmentation. In A549 cells, T/HS lymph activated caspase-3-mediated apoptosis, which was partially abrogated by N-benzyloxycarbonyl-Val-Ala-Asp (zVAD). Additionally, T/HS lymph did not cause the nuclear translocation of AIF in A549 cells. Collectively, T/HS-induced pulmonary endothelial PCD occurs via an AIF-dependent caspase-independent pathway, whereas epithelial cells undergo apoptosis by a caspase-dependent pathway.


Subject(s)
Acute Lung Injury/pathology , Apoptosis/physiology , Lung/pathology , Shock, Hemorrhagic/pathology , Acute Lung Injury/physiopathology , Animals , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/physiology , Caspases/physiology , Cells, Cultured , Endothelial Cells/pathology , Epithelial Cells/pathology , Humans , Lung/physiopathology , Lymph/physiology , Male , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/physiopathology , Signal Transduction
15.
Shock ; 30(6): 680-5, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18496238

ABSTRACT

The goal of this study was to test the hypothesis that factors released from the gut and carried in the mesenteric lymph contribute to mortality in a lethal gut I/R model. To test this hypothesis, a lethal splanchnic artery occlusion (SAO) shock model was used in male Sprague-Dawley rats. In the first set of experiments, ligation of the mesenteric lymph duct (LDL), which prevents gut-derived factors carried in the intestinal lymphatics from reaching the systemic circulation, significantly improved 24-h survival after a 20-min SAO insult (0% vs. 60% survival; P < 0.05). This increase in survival in the LDL-treated rats was associated with a blunted hypotensive response. Because increased iNOS-induced NO levels have been implicated in SAO-induced shock, we measured plasma nitrite/nitrate levels and liver iNOS protein levels in a second group of animals. Ligation of the mesenteric lymph duct significantly abrogated the SAO-induced increase in plasma nitrite/nitrate levels and the induction of hepatic iNOS (P < 0.05). In an additional series of studies, we documented that LDL increased not only 24-h but also long-term 7-day survival. During the course of these studies, we made the unexpected finding that Sprague-Dawley rats from different animal vendors had differential resistance to SAO, and that the time of the year that the experiments were carried out also influenced the results. Nonetheless, in conclusion, these studies support the hypothesis that factors carried in the mesenteric lymph significantly contribute to the development of irreversible shock after SAO.


Subject(s)
Ligation/methods , Lymphatic Vessels/surgery , Shock/mortality , Shock/surgery , Animals , Disease Models, Animal , Gastrointestinal Tract/immunology , Lymphatic Vessels/immunology , Male , Neutrophil Activation/physiology , Rats , Rats, Sprague-Dawley , Shock/immunology
16.
Ann Surg ; 246(5): 822-30, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17968175

ABSTRACT

OBJECTIVE: To test the hypothesis that gut-derived factors carried in trauma-hemorrhagic shock (T/HS) lymph is sufficient to induce lung injury. Additionally, because our previous studies showed that T/HS-induced nitric oxide production was associated with lung injury, we examined whether T/HS lymph-induced lung injury occurs via an inducible nitric oxide synthase (iNOS)-dependent pathway. BACKGROUND: We have previously shown that T/HS-induced lung injury is mediated by gut-derived humoral factors carried in the mesenteric lymph. However, it remains unclear whether T/HS lymph itself is sufficient to induce lung injury, or requires the activation of other factors during the T/HS period to exert its effect. METHODS: Mesenteric lymph collected from T/HS or trauma-sham shock (T/SS) animals was injected intravenously into male rats at a rate of 1 mL/h for 3 hours. At the end of infusion, lung injury was assessed by lung permeability and lung histology. The effect of iNOS inhibition on T/HS lymph-induced lung injury was studied and this was further confirmed in iNOS knockout mice. Finally, iNOS immunohistochemistry was performed to identify the cells of origin of iNOS. RESULTS: The injection of T/HS lymph, but not sham shock lymph, caused lung injury. This was associated with increased plasma nitrite/nitrate levels as well as induction of iNOS protein in the lung, liver, and gut. Treatment with the selective iNOS inhibitor aminoguanidine prevented T/HS lymph-induced lung injury. iNOS knockout mice, but not their wild-type controls, were resistant to T/HS lymph-induced lung injury. By immunohistochemistry, neutrophils and macrophages, rather than parenchymal cells, were the source of T/HS lymph-induced lung iNOS. CONCLUSIONS: These results indicate that T/HS lymph is sufficient to induce acute lung injury and that lymph-induced lung injury occurs via an iNOS-dependent pathway.


Subject(s)
Lymph/physiology , Nitric Oxide Synthase Type II/physiology , Respiratory Distress Syndrome/etiology , Shock, Hemorrhagic/complications , Animals , Disease Models, Animal , Injections, Intravenous , Male , Mesentery , Mice , Mice, Knockout , Nitric Oxide/physiology , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Shock, Hemorrhagic/metabolism
17.
Shock ; 28(3): 360-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17545943

ABSTRACT

The etiology of trauma-hemorrhagic shock (T/HS)-induced acute lung injury has been difficult to elucidate because of, at least in part, the inability of in vivo studies to separate the noninjurious pulmonary effects of trauma-hemorrhage from the tissue-injurious ones. To circumvent this in vivo limitation, we used a model of T/HS in which T/HS lung injury was abrogated by dividing the mesenteric lymph duct. In this way, it was possible to separate the pulmonary injurious response from the noninjurious systemic response to T/HS by comparing the pulmonary molecular responses of rats subjected to T/HS, which did and did not develop lung injury, with those of nonshocked rats. Using high-density oligonucleotide arrays and treatment group comparisons of whole lung tissue collected at 3 h after the end of the shock or sham-shock period, 139 of 8,799 assessed genes were identified by significant analysis of microarrays. Hemorrhage without the secondary effects of lung injury modulated the expression of 21 genes such as interleukin 1beta, metallothionein-2, and myeloctomatosis oncogene (c-myc). In response to injury, 42 genes were identified to be differentially expressed. Upregulated genes included the L1 retroposon and guanine deaminase, whereas downregulated genes included catalase and superoxide dismutase 1. Real-time polymerase chain reaction confirmed the differential expression for selected genes. PathwayAssist analysis identified interleukin 1beta as a central regulator of two subpathways of stress response-related genes (c-myc and superoxide dismutase 1/catalase) as well as several unrelated genes such as lipoprotein lipase. Our model system provided a unique opportunity to distinguish the molecular changes associated with T/HS-induced acute lung injury from the systemic molecular response to T/HS.


Subject(s)
Lung/metabolism , Respiratory Distress Syndrome/genetics , Shock, Hemorrhagic/genetics , Animals , Gene Expression Profiling , Ligation , Lymphatic Vessels , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/pathology
18.
Am J Physiol Gastrointest Liver Physiol ; 288(3): G466-72, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15499084

ABSTRACT

Having documented that proestrus female rats are more resistant to shock-induced acute gut and hence lung injury than male rats, we tested the hypothesis that the female gut is more resistant to injury and produces less of an inflammatory response than the male gut when exposed to conditions associated with shock states (hypoxia and acidosis) utilizing the ex vivo Ussing chamber system. Ileal mucosal membranes harvested from normal male and female rats mounted in Ussing chamber systems were exposed to normoxia or 40 min of hypoxia at a normal pH (pH 7.3) or acidosis (pH 6.8). Cytokine and nitric oxide levels in the serosal compartment of the Ussing chamber were measured at the end of the 3-h experimental period to assess the immunoinflammatory response, whereas FITC-dextran (mol wt 4,300) was employed to assess barrier function. Histomorphological changes were used to quantitate gut mucosal injury. Hypoxia, acidosis, or hypoxia plus acidosis was associated with a significant increase in proinflammatory cytokine production [interleukin (IL)-6, tumor necrosis factor, and macrophage inflammatory protein (MIP)-2] by the male compared with the female intestinal segments. In contrast, the female gut manifested a higher anti-inflammatory response (nitric oxide and IL-10) and improved intestinal barrier function as well as less evidence of mucosal injury than the male intestinal segments. Administration of estradiol or the testosterone receptor antagonist, flutamide, to male rats abrogated the increase in gut injury and the increased IL-6 and MIP-2 response observed after hypoxia plus acidosis. These results suggest that gender differences in the ex vivo intestinal response to stresses, such as hypoxia and acidosis, exist and that the administration of estradiol or blockade of the testosterone receptor to male rats mitigates these gender differences.


Subject(s)
Enteritis/physiopathology , Intestines/injuries , Intestines/physiopathology , Shock/physiopathology , Acidosis/physiopathology , Animals , Cell Membrane/metabolism , Chemokine CXCL2 , Diffusion Chambers, Culture , Enteritis/metabolism , Female , Hypoxia/physiopathology , Ileum/pathology , Ileum/physiopathology , In Vitro Techniques , Interleukin-10/metabolism , Interleukin-6/metabolism , Male , Monokines/metabolism , Multiple Organ Failure/physiopathology , Nitric Oxide/metabolism , Proestrus/physiology , Rats , Rats, Sprague-Dawley , Sex Characteristics , Shock/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Shock ; 22(3): 270-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15316398

ABSTRACT

In both animal models of hemorrhagic shock and clinical settings, shock-induced gut ischemia has been implicated in the development of the systemic inflammatory response syndrome and distant organ injury, yet the factors transducing these events remain to be fully determined. Because hypoxia-inducible factor (HIF-1), a transcription factor composed of oxygen-labile HIF-1alpha and constitutive HIF-1beta subunits, regulates the physiologic/pathophysiologic response to hypoxia and ischemia, we examined the HIF-1 response in two rat models of gut ischemia-reperfusion. We found that ileal nuclear HIF-1alpha protein levels were induced in rats subjected to trauma (laparotomy) plus hemorrhagic shock for 90 min relative to their trauma sham-shock and naïve counterparts and that this trauma hemorrhagic shock-induced mucosal HIF-1alpha protein response persisted after 1 h and 3 h of reperfusion. Likewise, in a model of isolated gut ischemia-reperfusion injury, where the superior mesenteric artery was occluded for 45 min, nuclear HIF-1alpha were induced in the gut mucosa relative to their sham counterparts and persisted after 1 h and 3 h or reperfusion. Similar to the in vivo response, in vitro hypoxia induced HIF-alpha expression in three different enterocyte cell lines (rat IEC-6 and human Caco-2 and HT-29 cell lines). However, in contrast to the in vivo response, HIF-1 expression rapidly disappeared on subsequent reoxygenation. Because in vivo enterocytes are exposed to bacteria, we tested whether the in vitro HIF-1alpha response would persist on reoxygenation if the enterocytes were cocultured with bacteria. P. aeruginosa, an enteric bacterium, markedly induced enterocyte HIF-1alpha protein levels under normoxic conditions. Furthermore, the addition of P. aeruginosa during either the hypoxic or reoxygenation phase prevented the degradation of HIF-1alpha protein levels. Moreover, the observation that lipopolysaccharide induced HIF-1alpha expression in a time-dependent manner in IEC-6 cells indicated that the induction of HIF-1 by exposure to P. aeruginosa is not dependent on bacterial viability. In conclusion, these results suggest that HIF-1alpha activation is an early reperfusion-independent event in models of gut ischemia-reperfusion and that this HIF-1alpha response is potentiated by the presence of P. aeruginosa or lipopolysaccharide.


Subject(s)
Intestines/blood supply , Lipopolysaccharides/toxicity , Reperfusion Injury/physiopathology , Shock, Hemorrhagic/physiopathology , Transcription Factors/physiology , Animals , Bacterial Infections , Cell Hypoxia , Cell Line , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/physiology , Colonic Neoplasms , Disease Models, Animal , Enterocytes/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Male , Plasmids , Pseudomonas aeruginosa , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics
20.
Clin Adv Hematol Oncol ; 2(3): 162-6, 2004 Mar.
Article in English | MEDLINE | ID: mdl-16166946

ABSTRACT

The nuclear factor kappa B (NF-kappaB) family of transcription factors plays a major role in inflammation, immune and stress responses, oncogenesis, cell migration, and angiogenesis. Aberrant activation of NF-kappaB has also been shown to contribute to intrinsic and inducible drug resistance in numerous cancers, including multiple myeloma. The expression of NF-kappaB-responsive targets will vary depending on the cellular context and type of inducer. The regulation of NF-kappaB activity occurs at multiple levels involving the IkappaB kinase (IKK) complex, members of the IkappaB family, recruitment of heterologous transcription factors and coactivators by NF-kappaB, and post-translational modifications of p65. This article highlights regulatory mechanisms responsible for constitutive NF-kappaB activation and provides justification for target-based therapy for NF-kappaB in multiple myeloma.


Subject(s)
Multiple Myeloma/drug therapy , Multiple Myeloma/physiopathology , NF-kappa B/physiology , Drug Resistance, Neoplasm , Humans , NF-kappa B/biosynthesis , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...