Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Genomics ; 24(1): 297, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37264318

ABSTRACT

BACKGROUND: Saussurea involucrata (Sik.) is alpine plant that have developed special adaptive mechanisms to resist adverse environmental conditions such as low temperature chilling during long-term adaptation and evolution. Exploring the changes of its metabolites under different temperature stresses is helpful to gain insight into its cold stress tolerance. METHODS: Ultra-performance liquid chromatography and tandem mass spectrometry were used to analyze the metabolites in the leaves of Sik. under low different temperature stress conditions. RESULTS: A total of 753 metabolites were identified, and 360 different metabolites were identified according to the Kyoto Encyclopedia of Genes and Genomes (KEGG) involved in the biosynthesis of secondary metabolites and amino acids and sugars. Sucrose and trehalose synthesis, glycolysis, tricarboxylic acid cycle, pentose phosphate pathway, glutamic acid-mediated proline biosynthesis, purine metabolism, amino acid metabolism, phenylpropane synthesis pathway metabolites all respond to low temperature stress. Under cold stress conditions, carbohydrates in Sik. leaves accumulate first than under freezing conditions, and the lower the temperature under freezing conditions, the less amino acids accumulate, while the phenolic substances increase. The expression of various substances in LPE and LPC increased more than 10-fold after low temperature stress compared with the control, but the content of LPE and LPC substances decreased after cold adaptation. In addition, purines and phenolics decreased and amino acids accumulated significantly under freezing conditions. CONCLUSION: The metabolic network of Sik. leaves under different low temperature stress conditions was proposed, which provided a reference for further exploration of the metabolic mechanism related to low temperature stress tolerance of Sik.


Subject(s)
Saussurea , Saussurea/genetics , Saussurea/metabolism , Temperature , Cold Temperature , Freezing , Metabolomics , Amino Acids/metabolism
2.
Small ; 18(44): e2204436, 2022 11.
Article in English | MEDLINE | ID: mdl-36098251

ABSTRACT

This study presents the first messenger RNA (mRNA) therapy for metastatic ovarian cancer and cachexia-induced muscle wasting based on lipid nanoparticles that deliver follistatin (FST) mRNA predominantly to cancer clusters following intraperitoneal administration. The secreted FST protein, endogenously synthesized from delivered mRNA, efficiently reduces elevated activin A levels associated with aggressive ovarian cancer and associated cachexia. By altering the cancer cell phenotype, mRNA treatment prevents malignant ascites, delays cancer progression, induces the formation of solid tumors, and preserves muscle mass in cancer-bearing mice by inhibiting negative regulators of muscle mass. Finally, mRNA therapy provides synergistic effects in combination with cisplatin, increasing the survival of mice and counteracting muscle atrophy induced by chemotherapy and cancer-associated cachexia. The treated mice develop few nonadherent tumors that are easily resected from the peritoneum. Clinically, this nanomedicine-based mRNA therapy can facilitate complete cytoreduction, target resistance, improve resilience during aggressive chemotherapy, and improve survival in advanced ovarian cancer.


Subject(s)
Nanoparticles , Ovarian Neoplasms , Humans , Female , Cachexia/drug therapy , Cachexia/metabolism , Follistatin/metabolism , Follistatin/pharmacology , Follistatin/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Ovarian Neoplasms/complications , Ovarian Neoplasms/therapy , Muscle, Skeletal/metabolism
3.
Brain Behav Immun ; 97: 102-118, 2021 10.
Article in English | MEDLINE | ID: mdl-34245812

ABSTRACT

Lipocalin 2 (LCN2) is a pleiotropic molecule that is induced in the central nervous system (CNS) in several acute and chronic pathologies. The acute induction of LCN2 evolved as a beneficial process, aimed at combating bacterial infection through the sequestration of iron from pathogens, while the role of LCN2 during chronic, non-infectious disease remains unclear, and recent studies suggest that LCN2 is neurotoxic. However, whether LCN2 is sufficient to induce behavioral and cognitive alterations remains unclear. In this paper, we sought to address the role of cerebral LCN2 on cognition in both acute and chronic settings. We demonstrate that LCN2 is robustly induced in the CNS during both acute and chronic inflammatory conditions, including LPS-based sepsis and cancer cachexia. In vivo, LPS challenge results in a global induction of LCN2 in the central nervous system, while cancer cachexia results in a distribution specific to the vasculature. Similar to these in vivo observations, in vitro modeling demonstrated that both glia and cerebral endothelium produce and secrete LCN2 when challenged with LPS, while only cerebral endothelium secrete LCN2 when challenged with cancer-conditioned medium. Chronic, but not short-term, cerebral LCN2 exposure resulted in reduced hippocampal neuron staining intensity, an increase in newborn neurons, microglial activation, and increased CNS immune cell infiltration, while gene set analyses suggested these effects were mediated through melanocortin-4 receptor independent mechanisms. RNA sequencing analyses of primary hippocampal neurons revealed a distinct transcriptome associated with prolonged LCN2 exposure, and ontology analysis was suggestive of altered neurite growth and abnormal spatial learning. Indeed, LCN2-treated hippocampal neurons display blunted neurite processes, and mice exposed to prolonged cerebral LCN2 levels experienced a reduction in spatial reference memory as indicated by Y-maze assessment. These findings implicate LCN2 as a pathologic mediator of cognitive decline in the setting of chronic disease.


Subject(s)
Cognitive Dysfunction , Neurons , Animals , Hippocampus/metabolism , Lipocalin-2 , Mice , Neuroglia/metabolism , Neurons/metabolism
4.
Glia ; 68(7): 1479-1494, 2020 07.
Article in English | MEDLINE | ID: mdl-32039522

ABSTRACT

Microglia in the mediobasal hypothalamus (MBH) respond to inflammatory stimuli and metabolic perturbations to mediate body composition. This concept is well studied in the context of high fat diet induced obesity (HFDO), yet has not been investigated in the context of cachexia, a devastating metabolic syndrome characterized by anorexia, fatigue, and muscle catabolism. We show that microglia accumulate specifically in the MBH early in pancreatic ductal adenocarcinoma (PDAC)-associated cachexia and assume an activated morphology. Furthermore, we observe astrogliosis in the MBH and hippocampus concurrent with cachexia initiation. We next show that circulating immune cells resembling macrophages infiltrate the MBH. PDAC-derived factors induced microglia to express a transcriptional profile in vitro that was distinct from that induced by lipopolysaccharide (LPS). Microglia depletion through CSF1-R antagonism resulted in accelerated cachexia onset and increased anorexia, fatigue, and muscle catabolism during PDAC. This corresponded with increased hypothalamic-pituitary-adrenal (HPA) axis activation. CSF1-R antagonism had little effect on inflammatory response in the circulation, liver, or tumor. These findings demonstrate that microglia are protective against PDAC cachexia and provide mechanistic insight into this function.


Subject(s)
Cachexia/metabolism , Hypothalamus/metabolism , Microglia/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cachexia/immunology , Energy Metabolism/physiology , Gliosis/metabolism , Inflammation/metabolism , Macrophages/metabolism , Mice , Obesity/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
5.
Brain Behav Immun ; 82: 338-353, 2019 11.
Article in English | MEDLINE | ID: mdl-31499172

ABSTRACT

Toll-like receptors 7 and 8 (TLR7 and TLR8) are endosomal pattern recognition receptors that detect a variety of single-stranded RNA species. While TLR7/8 agonists have robust therapeutic potential, clinical utility of these agents is limited by sickness responses associated with treatment induction. To understand the kinetics and mechanism of these responses, we characterized the acute and chronic effects of TLR7 stimulation. Single-cell RNA-sequencing studies, RNAscope, and radiolabeled in situ hybridization demonstrate that central nervous system gene expression of TLR7 is exclusive to microglia. In vitro studies demonstrate that microglia are highly sensitive to TLR7 stimulation, and respond in a dose-dependent manner to the imidazoquinoline R848. In vivo, both intraperitoneal (IP) and intracerebroventricular (ICV) R848 induce acute sickness responses including hypophagia, weight loss, and decreased voluntary locomotor activity, associated with increased CNS pro-inflammatory gene expression and changes to glial morphology. However, chronic daily IP R848 resulted in rapid tachyphylaxis of behavioral and molecular manifestations of illness. In microglial in vitro assays, pro-inflammatory transcriptional responses rapidly diminished in the context of repeated R848. In addition to TLR7 desensitization, we found that microglia become partially refractory to lipopolysaccharide (LPS) following R848 pretreatment, associated with induction of negative regulators A20 and Irak3. Similarly, mice pre-treated with R848 demonstrate reduced sickness responses, hypothalamic inflammation, and hepatic inflammation in response to LPS. These data combined demonstrate that TLR7 stimulation induces acute behavioral and molecular evidence of sickness responses. Following prolonged dosing, R848 induces a refractory state to both TLR7 and TLR4 activation, consistent with induced immune tolerance.


Subject(s)
Membrane Glycoproteins/agonists , Membrane Glycoproteins/immunology , Microglia/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/immunology , Animals , Behavior, Animal , Cells, Cultured , Central Nervous System/drug effects , Central Nervous System/immunology , Cytokines/immunology , Female , Imidazoles/pharmacology , Immune Tolerance/drug effects , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Microglia/drug effects , Signal Transduction/drug effects , Tachyphylaxis/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/immunology
6.
Brain Behav Immun ; 73: 364-374, 2018 10.
Article in English | MEDLINE | ID: mdl-29852290

ABSTRACT

Hypothalamic inflammation is a key component of acute sickness behavior and cachexia, yet mechanisms of inflammatory signaling in the central nervous system remain unclear. Previous work from our lab and others showed that while MyD88 is an important inflammatory signaling pathway for sickness behavior, MyD88 knockout (MyD88KO) mice still experience sickness behavior after inflammatory stimuli challenge. We found that after systemic lipopolysaccharide (LPS) challenge, MyD88KO mice showed elevated expression of several cytokine and chemokine genes in the hypothalamus. We therefore assessed the role of an additional inflammatory signaling pathway, TRIF, in acute inflammation (LPS challenge) and in a chronic inflammatory state (cancer cachexia). TRIFKO mice resisted anorexia and weight loss after peripheral (intraperitoneal, IP) or central (intracerebroventricular, ICV) LPS challenge and in a model of pancreatic cancer cachexia. Compared to WT mice, TRIFKO mice showed attenuated upregulation of Il6, Ccl2, Ccl5, Cxcl1, Cxcl2, and Cxcl10 in the hypothalamus after IP LPS treatment, as well as attenuated microglial activation and neutrophil infiltration into the brain after ICV LPS treatment. Lastly, we found that TRIF was required for Ccl2 upregulation in the hypothalamus and induction of the catabolic genes, Mafbx, Murf1, and Foxo1 in gastrocnemius during pancreatic cancer. In summary, our results show that TRIF is an important inflammatory signaling mediator of sickness behavior and cachexia and presents a novel therapeutic target for these conditions.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Cachexia/physiopathology , Illness Behavior/drug effects , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Brain/metabolism , Cytokines/metabolism , Female , Hypothalamus/metabolism , Illness Behavior/physiology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Neoplasms/metabolism , Signal Transduction/drug effects
7.
Transl Oncol ; 50: 102129, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39353236

ABSTRACT

BACKGROUND: The prognosis of pancreatic ductal adenocarcinomas (PDAC) remains very poor, emphasizing the critical importance of early detection, where biomarkers offer unique potential. Although growth differentiation factor 15 (GDF15) and Lipocalin 2 (LCN2) have been linked to PDAC, their precise roles as biomarkers are uncertain. METHODS: Circulating levels of GDF15 and LCN2 were examined in human PDAC patients, heathy controls, and individuals with benign pancreatic diseases. Circulating levels of IL-6, CA19-9, and neutrophil-to-lymphocyte ratio (NLR) were measured for comparisons. Correlations between PDAC progression and overall survival were assessed. A mouse PDAC model was employed for comprehensive analyses, complementing the human studies by exploring associations with various metabolic and inflammatory parameters. Sensitivity and specificity of the biomarkers were evaluated. FINDINGS: Our results demonstrated elevated levels of circulating GDF15 and LCN2 in PDAC patients compared to both healthy controls and individuals with benign pancreatic diseases, with higher GDF15 levels associated with disease progression and increased mortality. In PDAC mice, circulating GDF15 and LCN2 progressively increased, correlating with tumor growth, behavioral manifestations, tissue and molecular pathology, and cachexia development. GDF15 exhibited highly sensitive and specific for PDAC patients compared to CA19-9, IL-6, or NLR, while LCN2 showed even greater sensitivity and specificity in PDAC mice. Combining GDF15 and LCN2, or GDF15 and CA19-9, enhanced sensitivity and specificity. INTERPRETATION: Our findings indicate that GDF15 holds promise as a biomarker for early detection and prognosis of PDAC, while LCN2 could strengthen diagnostic panels.

8.
bioRxiv ; 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39386578

ABSTRACT

Tumor immune resistance is recognized as a contributor to low survivorship in pancreatic ductal adenocarcinoma (PDAC). We developed a novel murine model of spontaneous PDAC clearance, generated by overexpressing interleukin-6 (IL-6) in orthotopically implanted PDAC cancer cells (OT-PDAC IL6 ). Circulating IL-6 was 100-fold higher in OT-PDAC IL6 than in OT-PDAC parental mice. OT-PDAC IL6 tumors were present at 5 days post-implantation, and undetectable by 10 days post implantation. Flow cytometry revealed increased T cells and NK cells, and decreased T regulatory cells in OT-PDAC IL6 as compared to OT-PDAC parental tumors. Increased lymphoid aggregates were apparent by histological assessment and may account for elevated T cell content. Antibody-based depletion of CD4 + and CD8 + T cells prevented tumor clearance and significantly reduced survival of OT-PDAC IL6 mice. The anti-tumor immune response to OT-PDAC IL6 rendered mice immune to re-challenge with OT-PDAC parental tumors. In high concentrations, IL-6 acts in opposition to previously described pro-tumorigenic effects by enhancing the T cell-mediated anti-tumor response to PDAC. Statement of Significance: Interleukin 6 overexpression in pancreatic ductal adenocarcinoma cells induces T cell-driven tumor clearance that is rapid and durable. Supraphysiologic levels of interleukin 6 are sufficient to drive an anti-tumor immune microenvironment hallmarked by increased lymphoid aggregate formation, increased CD4 T cell abundance, and decreased Treg abundance.

9.
Plant Sci ; 332: 111704, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37037298

ABSTRACT

Drought is an important factor limiting the yield and quality of cotton. In the present study, the gene encoding the cotton calcium-dependent protein kinase GhCDPK4 was identified and characterized in the transcriptome of cotton under PEG-induced drought stress. In RT-qPCR experiments, GhCDPK4 expression was found to be up-regulated under drought and abscisic acid (ABA) stress. Under drought conditions, heterologous overexpression of GhCDPK4 in tobacco showed a better phenotypic status, higher antioxidant enzyme activity, and lower relative electrolyte leakage (REL) and malondialdehyde (MDA) content. Meanwhile, ghcdpk4-silenced cotton plants, which were extremely sensitive to drought, exhibited higher levels of O2-ĆÆĀ¼ĀŒH2O2, and MDA contents compared to the control. Meanwhile, silenced lines showed impaired stomatal closure under drought stress, resulting in increased water loss from transpiration in silenced lines. GhCDPK4 expression was induced by ABA, and there are five ABA-responsive elements in its promoter. and C2-DOMAIN ABA-RELATED 4(CAR4, Gh_D09G1653) were found to interact and be co-expressed in the GhCDPK4 interaction network. Therefore, GhCDPK4 may reduce the extent of water loss and oxidative damage in cotton under drought by positively regulating ABA-controlled stomatal closure and reactive oxygen species (ROS) scavenging systems. This study demonstrates the great potential of GhCDPK4 in improving drought resistance in crops.


Subject(s)
Abscisic Acid , Stress, Physiological , Abscisic Acid/metabolism , Stress, Physiological/genetics , Droughts , Hydrogen Peroxide/metabolism , Water/metabolism , Antioxidants/metabolism , Gene Expression Regulation, Plant , Plants, Genetically Modified/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism
10.
J Neurosci ; 31(31): 11376-86, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21813697

ABSTRACT

In response to illness, animals subvert normal homeostasis and divert their energy utilization to fight infection. An important and unexplored feature of this response is the suppression of physical activity and foraging behavior in the setting of negative energy balance. Inflammatory signaling in the hypothalamus mediates the febrile and anorectic responses to disease, but the mechanism by which locomotor activity (LMA) is suppressed has not been described. Lateral hypothalamic orexin (Ox) neurons link energy status with LMA, and deficiencies in Ox signaling lead to hypoactivity and hypophagia. In the present work, we examine the effect of endotoxin-induced inflammation on Ox neuron biology and LMA in rats. Our results demonstrate a vital role for diminished Ox signaling in mediating inflammation-induced lethargy. This work defines a specific population of inflammation-sensitive, arousal-associated Ox neurons and identifies a proximal neural target for inflammatory signaling to Ox neurons, while eliminating several others.


Subject(s)
Inflammation/complications , Intracellular Signaling Peptides and Proteins/metabolism , Lethargy/drug therapy , Lethargy/etiology , Neurons/metabolism , Neuropeptides/metabolism , Analysis of Variance , Animals , Dark Adaptation/drug effects , Disease Models, Animal , Drug Administration Routes , Enzyme-Linked Immunosorbent Assay/methods , Food Deprivation , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation/chemically induced , Injections, Intraventricular/methods , Interleukin-1beta/pharmacology , Interleukin-6/blood , Intracellular Signaling Peptides and Proteins/pharmacology , Lateral Ventricles/drug effects , Lateral Ventricles/physiology , Lethargy/pathology , Male , Melanocyte-Stimulating Hormones/pharmacology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , NF-KappaB Inhibitor alpha , Neoplasm Transplantation/methods , Neurons/drug effects , Neuropeptides/pharmacology , Neurotensin/genetics , Orexins , Photoperiod , Polysaccharides/adverse effects , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Corticotropin/antagonists & inhibitors , Receptors, Interleukin-11/genetics , Receptors, Interleukin-11/metabolism , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/metabolism
11.
Am J Physiol Endocrinol Metab ; 303(12): E1446-58, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23047987

ABSTRACT

Animals exhibit a rapid and sustained anorexia when fed a diet that is deficient in a single indispensable amino acid (IAA). The chemosensor for IAA deficiency resides within the anterior piriform cortex (APC). Although the cellular and molecular mechanisms by which the APC detects IAA deficiency are well established, the efferent neural pathways that reduce feeding in response to an IAA-deficient diet remain to be fully characterized. In the present work, we investigated whether 1) central melanocortin signaling is involved in IAA deficiency-induced anorexia (IAADA) and 2) IAADA engages other key appetite-regulating neuronal populations in the hypothalamus. Rats and mice that consumed a valine-deficient diet (VDD) for 2-3 wk exhibited marked reductions in food intake, body weight, fat and lean body mass, body temperature, and white adipose tissue leptin gene expression, as well as a paradoxical increase in brown adipose tissue uncoupling protein-1 mRNA. Animals consuming the VDD had altered hypothalamic gene expression, typical of starvation. Pharmacological and genetic blockade of central melanocortin signaling failed to increase long-term food intake in this model. Chronic IAA deficiency was associated with a marked upregulation of corticotropin-releasing hormone expression in the lateral hypothalamus, particularly in the parasubthalamic nucleus, an area heavily innervated by efferent projections from the APC. Our observations indicate that the hypothalamic melanocortin system plays a minor role in acute, but not chronic, IAADA and suggest that the restraint on feeding is analogous to that observed after chronic dehydration.


Subject(s)
Anorexia/etiology , Anorexia/metabolism , Hypothalamus/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Signal Transduction , Valine/deficiency , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Anorexia/pathology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Female , Gene Expression Regulation , Hypothalamus/pathology , Ion Channels/genetics , Ion Channels/metabolism , Leptin/genetics , Leptin/metabolism , Male , Melanocortins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neural Pathways/pathology , Neurons/pathology , Organ Specificity , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Uncoupling Protein 1 , Valine/metabolism
12.
PeerJ ; 10: e12883, 2022.
Article in English | MEDLINE | ID: mdl-35186477

ABSTRACT

BACKGROUND: Calcium dependent protein kinases (CDPKs) are a class of important calcium signal sensing response proteins, which play an important regulatory role in response to abiotic stress. However, researchers have not been excavated CDPKs' role in drought in sea-island cotton(Gossypium barbadense L. 'H7124'). RESULTS: Eighty-four CDPK genes have been identified in G. barbadense. These GbCDPK genes are unevenly distributed on 26 chromosomes, and segmental duplication is the significant way for the extension of CDPK family. Also, members within the same subfamily share a similar gene structure and motif composition. There are a large number of cis-elements involved in plant growth and response to stresses in the promoter regions of GbCDPKs. Additionally, these GbCDPKs show differential expression patterns in cotton tissues. The transcription levels of most genes were markedly altered in cotton under heat, cold, salt and PEG treatments, while the expressions of some GbCDPKs were induced in cotton under drought stress. Among these drought-induced genes, we selected GbCDPK32, GbCDPK68, GbCDPK74, GbCDPK80 and GbCDPK83 for further functional characterization by virus-induced gene silencing (VIGS) method. CONCLUSIONS: In conclusion, the principal findings of this prospective study are that CDPKs were associated with drought. These findings provide a solid foundation for the development of future molecular mechanism in sea-island cotton.


Subject(s)
Droughts , Gossypium , Gossypium/genetics , Calcium , Prospective Studies , Genome, Plant/genetics
13.
J Cachexia Sarcopenia Muscle ; 13(6): 3014-3027, 2022 12.
Article in English | MEDLINE | ID: mdl-36303458

ABSTRACT

BACKGROUND: Cachexia-anorexia syndrome is a complex metabolic condition characterized by skeletal muscle wasting, reduced food intake and prominent involvement of systemic and central inflammation. Here, the gut barrier function was investigated in pancreatic cancer-induced cachexia mouse models by relating intestinal permeability to the degree of cachexia. We further investigated the involvement of the gut-brain axis and the crosstalk between tumour, gut and hypothalamus in vitro. METHODS: Two distinct mouse models of pancreatic cancer cachexia (KPC and 4662) were used. Intestinal inflammation and permeability were assessed through fluorescein isothiocyanate dextran (FITC-dextran) and lipopolysaccharide (LPS), and hypothalamic and systemic inflammation through mRNA expression and plasma cytokines, respectively. To simulate the tumour-gut-brain crosstalk, hypothalamic (HypoE-N46) cells were incubated with cachexia-inducing tumour secretomes and LPS. A synthetic mimic of C26 secretome was produced based on its secreted inflammatory mediators. Each component of the mimic was systematically omitted to narrow down the key mediator(s) with an amplifying inflammation. To substantiate its contribution, cyclooxygenase-2 (COX-2) inhibitor was used. RESULTS: In vivo experiments showed FITC-dextran was enhanced in the KPC group (362.3 vs. sham 111.4Ā ng/mL, PĀ <Ā 0.001). LPS was increased to 140.9Ā ng/mL in the KPC group, compared with sham and 4662 groups (115.8 and 115.8Ā ng/mL, PĀ <Ā 0.05). Hypothalamic inflammatory gene expression of Ccl2 was up-regulated in the KPC group (6.3 vs. sham 1, PĀ <Ā 0.0001, 4662 1.3, PĀ <Ā 0.001), which significantly correlated with LPS concentration (rĀ =Ā 0.4948, PĀ =Ā 0.0226). These data suggest that intestinal permeability is positively related to the cachexic degree. Prostaglandin E2 (PGE2) was confirmed to be present in the plasma and PGE2 concentration (log10) in the KPC group was much higher than in 4662 group (1.85 and 0.56Ā ng/mL, PĀ <Ā 0.001), indicating a role for PGE2 in pancreatic cancer-induced cachexia. Parallel to in vivo findings, in vitro experiments revealed that the cachexia-inducing tumour secretomes (C26, LLC, KPC and 4662) amplified LPS-induced hypothalamic IL-6 secretion (419%, 321%, 294%, 160%). COX-2 inhibitor to the tumour cells reduced PGE2 content (from 105 to 102 Ā pg/mL) in the secretomes and eliminated the amplified hypothalamic IL-6 production. Moreover, results could be reproduced by addition of PGE2 alone, indicating that the increased hypothalamic inflammation is directly related to the PGE2 from tumour. CONCLUSIONS: PGE2 secreted by the tumour may play a role in amplifying the effects of bacteria-derived LPS on the inflammatory hypothalamic response. The cachexia-inducing potential of tumour mice models parallels the loss of intestinal barrier function. Tumour-derived PGE2 might play a key role in cancer-related cachexia-anorexia syndrome via tumour-gut-brain crosstalk.


Subject(s)
Dinoprostone , Pancreatic Neoplasms , Animals , Mice , Anorexia , Anti-Inflammatory Agents, Non-Steroidal , Cachexia/pathology , Cyclooxygenase Inhibitors , Disease Models, Animal , Inflammation/metabolism , Interleukin-6 , Lipopolysaccharides , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
14.
J Cachexia Sarcopenia Muscle ; 12(5): 1312-1332, 2021 10.
Article in English | MEDLINE | ID: mdl-34231343

ABSTRACT

BACKGROUND: Cancer cachexia is a metabolic disorder characterized by the progressive loss of fat and lean mass that results in significant wasting, ultimately leading to reduced quality of life and increased mortality. Effective therapies for cachexia are lacking, potentially owing to the mismatch in clinically relevant models of cachexia. Specifically, cachexia observed in a clinical setting is commonly associated with advanced or late-stage cancers that are metastatic, yet pre-clinical metastatic models of cachexia are limited. Furthermore, the prevalence of cachexia in head and neck cancer patients is high, yet few pre-clinical models of head and neck cancer cachexia exist. In addition to these shortcomings, cachexia is also heterogeneous among any given cancer, whereas patients with similar disease burden may experience significantly different degrees of cachexia symptoms. In order to address these issues, we characterize a metastatic model of human papilloma virus (HPV) positive head and neck squamous cell carcinoma that recapitulates the cardinal clinical and molecular features of cancer cachexia. METHODS: Male and female C57BL/6 mice were implanted subcutaneously with oropharyngeal squamous cell carcinoma cells stably transformed with HPV16 E6 and E7 together with hRas and luciferase (mEERL) that metastasizes to the lungs (MLM). We then robustly characterize the physiologic, behavioural, and molecular signatures during tumour development in two MLM subclones. RESULTS: Mice injected with MLM tumour cells rapidly developed primary tumours and eventual metastatic lesions to the lungs. MLM3, but not MLM5, engrafted mice progressively lost fat and lean mass during tumour development despite the absence of anorexia (PĀ <Ā 0.05). Behaviourally, MLM3-implanted mice displayed decreased locomotor behaviours and impaired nest building (PĀ <Ā 0.05). Muscle catabolism programmes associated with cachexia, including E3 ubiquitin ligase and autophagy up-regulation, along with progressive adipose wasting and accompanying browning gene signatures, were observed. Tumour progression also corresponded with hypothalamic and peripheral organ inflammation, as well as an elevation in neutrophil-to-lymphocyte ratio (PĀ <Ā 0.05). Finally, we characterize the fat and lean mass sparing effects of voluntary wheel running on MLM3 cachexia (PĀ <Ā 0.05). CONCLUSIONS: This syngeneic MLM3 allograft model of metastatic cancer cachexia is reliable, consistent, and readily recapitulates key clinical and molecular features and heterogeneity of cancer cachexia. Because few metastatic models of cachexia exist-even though cachexia often accompanies metastatic progression-we believe this model more accurately captures cancer cachexia observed in a clinical setting and thus is well suited for future mechanistic studies and pre-clinical therapy development for this crippling metabolic disorder.


Subject(s)
Cachexia , Head and Neck Neoplasms , Animals , Cachexia/etiology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Motor Activity , Quality of Life
15.
Nat Commun ; 12(1): 2057, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33824339

ABSTRACT

Lipocalin 2 (LCN2) was recently identified as an endogenous ligand of the type 4 melanocortin receptor (MC4R), a critical regulator of appetite. However, it remains unknown if this molecule influences appetite during cancer cachexia, a devastating clinical entity characterized by decreased nutrition and progressive wasting. We demonstrate that LCN2 is robustly upregulated in murine models of pancreatic cancer, its expression is associated with reduced food consumption, and Lcn2 deletion is protective from cachexia-anorexia. Consistent with LCN2's proposed MC4R-dependent role in cancer-induced anorexia, pharmacologic MC4R antagonism mitigates cachexia-anorexia, while restoration of Lcn2 expression in the bone marrow is sufficient in restoring the anorexia feature of cachexia. Finally, we observe that LCN2 levels correlate with fat and lean mass wasting and is associated with increased mortality in patients with pancreatic cancer. Taken together, these findings implicate LCN2 as a pathologic mediator of appetite suppression during pancreatic cancer cachexia.


Subject(s)
Appetite , Cachexia/complications , Lipocalin-2/metabolism , Pancreatic Neoplasms/complications , Adult , Aged , Aged, 80 and over , Animals , Anorexia/blood , Anorexia/complications , Blood-Brain Barrier/pathology , Bone Marrow/pathology , Cachexia/blood , Cell Line, Tumor , Disease Models, Animal , Feeding Behavior , Female , Gene Deletion , Humans , Lipocalin-2/blood , Male , Mice, Knockout , Middle Aged , Models, Biological , Muscles/pathology , Neutrophils/pathology , Organ Size , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/genetics , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/metabolism , Up-Regulation
16.
Kidney Int ; 77(1): 23-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19890275

ABSTRACT

Skeletal muscle mitochondrial dysfunction and insulin resistance occur in chronic kidney disease. Ghrelin is a gastric hormone previously shown to enhance muscle mitochondrial enzyme activities and AKT-mediated insulin signaling independent of food intake in healthy rats. Here we determined the impact of ghrelin treatment on anorexia, skeletal muscle mitochondrial oxidative capacity, AKT phosphorylation as a measure of insulin signaling, and lean body mass in a rat model of chronic kidney disease. Ghrelin infusion promoted higher food intake and lean body mass. Further, although muscle mitochondrial enzyme activities were low in the rats with CKD (chronic kidney disease), they normalized with ghrelin treatment, a change that was consistent with the increase in the transcript levels of regulators of mitochondrial biogenesis and lipid metabolism. This was associated with a lower muscle triglyceride content and higher AKT phosphorylation. Pair-feeding showed that mitochondrial effects of ghrelin are independent of changes in food intake, whereas combined ghrelin treatment and higher food intake were needed to enhance AKT phosphorylation. Thus, ghrelin-induced muscle mitochondrial changes and lower tissue triglycerides could favor insulin action and muscle anabolism in the presence of improvement in food intake. Our study shows that combined effects of ghrelin on appetite and muscle mitochondria improve muscle metabolic and nutritional alterations in chronic kidney disease. This could have potential beneficial impact on patient morbidity and survival.


Subject(s)
Eating/drug effects , Ghrelin/pharmacology , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Insulin , Kidney Failure, Chronic , Mitochondria, Muscle/enzymology , Muscle, Skeletal/ultrastructure , Nutritional Status , Oxidation-Reduction , Phosphorylation , Rats , Triglycerides
17.
J Clin Invest ; 130(9): 4921-4934, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32544087

ABSTRACT

Cachexia, a devastating wasting syndrome characterized by severe weight loss with specific losses of muscle and adipose tissue, is driven by reduced food intake, increased energy expenditure, excess catabolism, and inflammation. Cachexia is associated with poor prognosis and high mortality and frequently occurs in patients with cancer, chronic kidney disease, infection, and many other illnesses. There is no effective treatment for this condition. Hypothalamic melanocortins have a potent and long-lasting inhibitory effect on feeding and anabolism, and pathophysiological processes increase melanocortin signaling tone, leading to anorexia, metabolic changes, and eventual cachexia. We used 3 rat models of anorexia and cachexia (LPS, methylcholanthrene sarcoma, and 5/6 subtotal nephrectomy) to evaluate efficacy of TCMCB07, a synthetic antagonist of the melanocortin-4 receptor. Our data show that peripheral treatment using TCMCB07 with intraperitoneal, subcutaneous, and oral administration increased food intake and body weight and preserved fat mass and lean mass during cachexia and LPS-induced anorexia. Furthermore, administration of TCMCB07 diminished hypothalamic inflammatory gene expression in cancer cachexia. These results suggest that peripheral TCMCB07 treatment effectively inhibits central melanocortin signaling and therefore stimulates appetite and enhances anabolism, indicating that TCMCB07 is a promising drug candidate for treating cachexia.


Subject(s)
Cachexia/drug therapy , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Renal Insufficiency, Chronic/drug therapy , Sarcoma, Experimental/drug therapy , Animals , Appetite/drug effects , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Male , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 4/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Sarcoma, Experimental/complications , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology
18.
Elife ; 92020 05 11.
Article in English | MEDLINE | ID: mdl-32391790

ABSTRACT

Weight loss and anorexia are common symptoms in cancer patients that occur prior to initiation of cancer therapy. Inflammation in the brain is a driver of these symptoms, yet cellular sources of neuroinflammation during malignancy are unknown. In a mouse model of pancreatic ductal adenocarcinoma (PDAC), we observed early and robust myeloid cell infiltration into the brain. Infiltrating immune cells were predominately neutrophils, which accumulated at a unique central nervous system entry portal called the velum interpositum, where they expressed CCR2. Pharmacologic CCR2 blockade and genetic deletion of Ccr2 both resulted in significantly decreased brain-infiltrating myeloid cells as well as attenuated cachexia during PDAC. Lastly, intracerebroventricular blockade of the purinergic receptor P2RX7 during PDAC abolished immune cell recruitment to the brain and attenuated anorexia. Our data demonstrate a novel function for the CCR2/CCL2 axis in recruiting neutrophils to the brain, which drives anorexia and muscle catabolism.


Weight loss, decreased appetite and fatigue are symptoms of a wasting disorder known as cachexia, which is common in several serious diseases such as AIDS, chronic lung disease and heart failure. Up to 80 percent of people with advanced cancer also develop cachexia, and there are no effective treatments. It is not known how cachexia develops, but symptoms like appetite loss and fatigue are controlled by the brain. One theory is that the brain may be responding to a malfunctioning immune response that causes inflammation. While the brain was thought to be protected from this, new research has shown that it is possible for cells from the immune system to reach the brain in some conditions. To find out if this also happens in cancer, Burfeind et al. studied mice that had been implanted with pancreatic cancer cells and were showing signs of cachexia. Samples from the mice's brains showed that immune cells known as neutrophils were present and active. A protein known as CCR2 was found in higher levels in the brains of these mice. This protein is involved in the movement of neutrophil cells through the body. To see what effect this protein had, Burfeind et al. gave the mice a drug that blocks CCR2. This prevented the neutrophils from entering the brain and reduced the symptoms of cachexia in the mice. To further confirm the role of CCR2, the mice were genetically modified so that they could not produce the protein. This reduced the number of neutrophils seen in the brain but not in the rest of the body. This suggests that a drug targeting CCR2 could help to reduce the symptoms of cachexia, without disrupting the normal immune response away from the brain. This approach would still need to be tested in clinical trials before it is possible to know how effective it might be in humans.


Subject(s)
Brain/physiopathology , Cachexia/etiology , Carcinoma, Pancreatic Ductal/pathology , Myeloid Cells/metabolism , Pancreatic Neoplasms/pathology , Animals , Anorexia/etiology , Carcinoma, Pancreatic Ductal/complications , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Disease Models, Animal , Female , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/immunology , Neutrophil Infiltration , Neutrophils/metabolism , Pancreatic Neoplasms/complications , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Weight Loss
19.
JAMA Otolaryngol Head Neck Surg ; 146(8): 714-722, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32525518

ABSTRACT

Importance: The negative association of low lean muscle mass (sarcopenia) with survival outcomes in head and neck cancers, including oropharyngeal carcinoma, is established. However, it is not known whether the choice of primary treatment modality (surgery or radiotherapy) is associated with oncologic outcomes of patients with sarcopenia and oropharyngeal squamous cell carcinoma (OPSCC). Objective: To examine whether primary surgical resection or definitive radiotherapy is associated with improved survival for patients with sarcopenia and localized OPSCC. Design, Setting, and Participants: A cohort study was conducted of patients with clinically staged T1 to T2, N0 to N2 OPSCC with cross-sectional abdominal imaging within 60 days prior to treatment and treated between January 1, 2005, and December 31, 2017. Skeletal muscle mass was measured at the third lumbar vertebra using previously defined techniques and sarcopenia was defined as less than 52.4 cm2/m2 of muscle for men and less than 38.5 cm2/m2 for women. In addition, associated patient demographic characteristics, cancer data, treatment information, and survival outcomes were assessed. Statistical analysis was performed from December 3, 2018, to August 28, 2019. Main Outcomes and Measures: Primary outcomes were overall survival and disease-specific survival. Results: Among the 245 patients who met study inclusion criteria, 209 were men (85.3%) and the mean (SD) age was 62.3 (7.8) years. Sarcopenia was detected in 135 patients (55.1%), while normal skeletal muscle mass was detected in 110 patients (44.9%). For the 110 patients without sarcopenia, primary treatment modality was not associated with improved survival. For patients with sarcopenia at diagnosis, primary surgical resection was associated with improved overall survival (hazard ratio [HR], 0.37; 95% CI, 0.17-0.82) and disease-specific survival (HR, 0.22; 95% CI, 0.07-0.68). This association persisted after propensity score matching, as up-front surgery was associated with improved overall survival (HR, 0.33; 95% CI, 0.12-0.91) and disease-specific survival (HR, 0.17; 95% CI, 0.04-0.75) survival. Conclusions and Relevance: This study suggests that sarcopenia has a negative association with survival for patients with OPSCC. Primary surgery and radiotherapy confer similar survival associations for patients with normal skeletal muscle mass and localized OPSCC. However, up-front surgical resection may be associated with improved survival outcomes for patients with sarcopenia.


Subject(s)
Head and Neck Neoplasms/therapy , Propensity Score , Robotic Surgical Procedures/methods , Sarcopenia/etiology , Squamous Cell Carcinoma of Head and Neck/therapy , Aged , Cross-Sectional Studies , Female , Follow-Up Studies , Head and Neck Neoplasms/complications , Head and Neck Neoplasms/diagnosis , Humans , Incidence , Male , Middle Aged , Neoplasm Staging , Oregon/epidemiology , Retrospective Studies , Sarcopenia/diagnosis , Sarcopenia/epidemiology , Squamous Cell Carcinoma of Head and Neck/complications , Squamous Cell Carcinoma of Head and Neck/diagnosis , Survival Rate/trends
20.
J Comp Neurol ; 527(17): 2826-2842, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31045239

ABSTRACT

When energy balance is altered by aerobic exercise, starvation, and cold exposure, for example, there appears to be coordination of the responses of skeletal muscle, white adipose (WAT), and brown adipose (BAT) tissues. We hypothesized that WAT, BAT, and skeletal muscle may share an integrated regulation by the central nervous system (CNS); specifically, that neurons in brain regions associated with energy balance would possess neuroanatomical connections to permit coordination of multiple, complementary responses in these downstream tissues. To study this, we used trans-neuronal viral retrograde tract tracing, using isogenic strains of pseudorabies virus (PRV) with distinct fluorescent reporters (either eGFP or mRFP), injected pairwise into male rat gastrocnemius, subcutaneous WAT and interscapular BAT, coupled with neurochemical characterization of specific cell populations for cocaine- and amphetamine-related transcript (CART), oxytocin (OX), corticotrophin releasing hormone (CRH) and calcitonin gene-related peptide (CGRP). Cells in the paraventricular (PVN) and parabrachial (PBN) nuclei and brainstem showed dual projections to muscle + WAT, muscle + BAT, and WAT + BAT. Dual PRV-labeled cells were found in parvocellular, magnocellular and descending/pre-autonomic regions of the PVN, and multiple structural divisions of the PBN and brainstem. In most PBN subdivisions, more than 50% of CGRP cells dually projected to muscle + WAT and muscle + BAT. Similarly, 31-68% of CGRP cells projected both to WAT + BAT. However, dual PRV-labeled cells in PVN only occasionally expressed OX or CRH but not CART. These studies reveal for the first time both separate and shared outflow circuitries among skeletal muscle and subcutaneous WAT and BAT.


Subject(s)
Adipose Tissue, Brown/innervation , Adipose Tissue, White/innervation , Brain Stem/cytology , Muscle, Skeletal/innervation , Neurons/cytology , Paraventricular Hypothalamic Nucleus/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Brain Stem/metabolism , Male , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Neural Pathways/cytology , Neural Pathways/metabolism , Neuroanatomical Tract-Tracing Techniques , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL