Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Small ; 20(4): e2306270, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37702136

ABSTRACT

Persistent and uncontrolled inflammation is the root cause of various debilitating diseases. Given that interleukin-1 receptor-associated kinase 4 (IRAK4) is a critical modulator of inflammation, inhibition of its activity with selective drug molecules (IRAK4 inhibitors) represents a promising therapeutic strategy for inflammatory disorders. To exploit the full potential of this treatment approach, drug carriers for efficient delivery of IRAK4 inhibitors to inflamed tissues are essential. Herein, the first nanoparticle-based platform for the targeted systemic delivery of a clinically tested IRAK4 inhibitor, PF-06650833, with limited aqueous solubility (57 µg mL-1 ) is presented. The developed nanocarriers increase the intrinsic aqueous dispersibility of this IRAK4 inhibitor by 40 times. A targeting peptide on the surface of nanocarriers significantly enhances their accumulation after intravenous injection in inflamed tissues of mice with induced paw edema and ulcerative colitis when compared to non-targeted counterparts. The delivered IRAK4 inhibitor markedly abates inflammation and dramatically suppresses paw edema, mitigates colitis symptoms, and reduces proinflammatory cytokine levels in the affected tissues. Importantly, repeated injections of IRAK4 inhibitor-loaded nanocarriers have no acute toxic effect on major organs of mice. Therefore, the developed nanocarriers have the potential to significantly improve the therapeutic efficacy of IRAK4 inhibitors for different inflammatory diseases.


Subject(s)
Colitis , Interleukin-1 Receptor-Associated Kinases , Mice , Animals , Interleukin-1 Receptor-Associated Kinases/chemistry , Cytokines , Inflammation/drug therapy , Edema
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.
Mol Metab ; 58: 101441, 2022 04.
Article in English | MEDLINE | ID: mdl-35031523

ABSTRACT

OBJECTIVE: Cancer cachexia is a devastating chronic condition characterized by involuntary weight loss, muscle wasting, abnormal fat metabolism, anorexia, and fatigue. However, the molecular mechanisms underlying this syndrome remain poorly understood. In particular, the hypothalamus may play a central role in cachexia, given that it has direct access to peripheral signals because of its anatomical location and attenuated blood-brain barrier. Furthermore, this region has a critical role in regulating appetite and metabolism. METHODS: To provide a detailed analysis of the hypothalamic response to cachexia, we performed single-cell RNA-seq combined with RNA-seq of the medial basal hypothalamus (MBH) in a mouse model for pancreatic cancer. RESULTS: We found many cell type-specific changes, such as inflamed endothelial cells, stressed oligodendrocyes and both inflammatory and moderating microglia. Lcn2, a newly discovered hunger suppressing hormone, was the highest induced gene. Interestingly, cerebral treatment with LCN2 not only induced many of the observed molecular changes in cachexia but also affected gene expression in food-intake decreasing POMC neurons. In addition, we found that many of the cachexia-induced molecular changes found in the hypothalamus mimic those at the primary tumor site. CONCLUSION: Our data reveal that multiple cell types in the MBH are affected by tumor-derived factors or host factors that are induced by tumor growth, leading to a marked change in the microenvironment of neurons critical for behavioral, metabolic, and neuroendocrine outputs dysregulated during cachexia. The mechanistic insights provided in this study explain many of the clinical features of cachexia and will be useful for future therapeutic development.


Subject(s)
Cachexia , Pancreatic Neoplasms , Animals , Cachexia/metabolism , Endothelial Cells/metabolism , Gene Regulatory Networks , Hypothalamus/metabolism , Mice , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Sequence Analysis, RNA , Tumor Microenvironment , Pancreatic Neoplasms
4.
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 (P < 0.05). Behaviourally, MLM3-implanted mice displayed decreased locomotor behaviours and impaired nest building (P < 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 (P < 0.05). Finally, we characterize the fat and lean mass sparing effects of voluntary wheel running on MLM3 cachexia (P < 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
5.
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
6.
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
7.
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
8.
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
10.
Nat Commun ; 10(1): 4682, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31615993

ABSTRACT

A priority in cancer research is to innovate therapies that are not only effective against tumor progression but also address comorbidities such as cachexia that limit quality and quantity of life. We demonstrate that TLR7/8 agonist R848 induces anti-tumor responses and attenuates cachexia in murine models of pancreatic ductal adenocarcinoma (PDAC). In vivo, tumors from two of three cell lines were R848-sensitive, resulting in smaller tumor mass, increased immune complexity, increased CD8+ T-cell infiltration and activity, and decreased Treg frequency. R848-treated mice demonstrated improvements in behavioral and molecular cachexia manifestations, resulting in a near-doubling of survival duration. Knockout mouse studies revealed that stromal, not neoplastic, TLR7 is requisite for R848-mediated responses. In patient samples, we found Tlr7 is ubiquitously expressed in stroma across all stages of pancreatic neoplasia, but epithelial Tlr7 expression is relatively uncommon. These studies indicate immune-enhancing approaches including R848 may be useful in PDAC and cancer-associated cachexia.


Subject(s)
Cachexia , Carcinoma, Pancreatic Ductal/metabolism , Imidazoles/pharmacology , Pancreatic Intraductal Neoplasms/metabolism , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/drug effects , Animals , Body Weight/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Eating/drug effects , Gene Expression , Humans , Locomotion/drug effects , Mice , Mice, Knockout , Pancreatic Intraductal Neoplasms/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Sequence Analysis, RNA , Survival Rate , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/agonists , Tumor Burden , Tumor Microenvironment/immunology
11.
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
12.
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
13.
J Cachexia Sarcopenia Muscle ; 8(5): 824-838, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28730707

ABSTRACT

BACKGROUND: Cachexia is a complex metabolic and behavioural syndrome lacking effective therapies. Pancreatic ductal adenocarcinoma (PDAC) is one of the most important conditions associated with cachexia, with >80% of PDAC patients suffering from the condition. To establish the cardinal features of a murine model of PDAC-associated cachexia, we characterized the effects of implanting a pancreatic tumour cell line from a syngeneic C57BL/6 KRASG12D P53R172H Pdx-Cre+/+ (KPC) mouse. METHODS: Male and female C57BL/6 mice were inoculated subcutaneously, intraperitoneally, or orthotopically with KPC tumour cells. We performed rigorous phenotypic, metabolic, and behavioural analysis of animals over the course of tumour development. RESULTS: All routes of administration produced rapidly growing tumours histologically consistent with moderate to poorly differentiated PDAC. The phenotype of this model was dependent on route of administration, with orthotopic and intraperitoneal implantation inducing more severe cachexia than subcutaneous implantation. KPC tumour growth decreased food intake, decreased adiposity and lean body mass, and decreased locomotor activity. Muscle catabolism was observed in both skeletal and cardiac muscles, but the dominant catabolic pathway differed between these tissues. The wasting syndrome in this model was accompanied by hypothalamic inflammation, progressively decreasing brown and white adipose tissue uncoupling protein 1 (Ucp1) expression, and increased peripheral inflammation. Haematological and endocrine abnormalities included neutrophil-dominant leukocytosis and anaemia, and decreased serum testosterone. CONCLUSIONS: Syngeneic KPC allografts are a robust model for studying cachexia, which recapitulate key features of the PDAC disease process and induce a wide array of cachexia manifestations. This model is therefore ideally suited for future studies exploring the physiological systems involved in cachexia and for preclinical studies of novel therapies.


Subject(s)
Cachexia/etiology , Cachexia/pathology , Pancreatic Neoplasms/complications , Allografts , Anemia/etiology , Anemia/metabolism , Anemia/pathology , Animals , Biopsy , Body Composition , Cachexia/diagnostic imaging , Disease Models, Animal , Energy Metabolism , Female , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Leukocytosis , Locomotion , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Muscles/metabolism , Muscles/pathology , Neutrophil Infiltration , Testosterone/metabolism
14.
J Neuroinflammation ; 14(1): 133, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28668091

ABSTRACT

BACKGROUND: During acute infections and chronic illnesses, the pro-inflammatory cytokine interleukin-1ß (IL-1ß) acts within the brain to elicit metabolic derangements and sickness behaviors. It is unknown which cells in the brain are the proximal targets for IL-1ß with respect to the generation of these illness responses. We performed a series of in vitro experiments to (1) investigate which brain cell populations exhibit inflammatory responses to IL-1ß and (2) examine the interactions between different IL-1ß-responsive cell types in various co-culture combinations. METHODS: We treated primary cultures of murine brain microvessel endothelial cells (BMEC), astrocytes, and microglia with PBS or IL-1ß, and then performed qPCR to measure inflammatory gene expression or immunocytochemistry to evaluate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. To evaluate whether astrocytes and/or BMEC propagate inflammatory signals to microglia, we exposed microglia to astrocyte-conditioned media and co-cultured endothelial cells and glia in transwells. Treatment groups were compared by Student's t tests or by ANOVA followed by Bonferroni-corrected t tests. RESULTS: IL-1ß increased inflammatory gene expression and NF-κB activation in primary murine-mixed glia, enriched astrocyte, and BMEC cultures. Although IL-1ß elicited minimal changes in inflammatory gene expression and did not induce the nuclear translocation of NF-κB in isolated microglia, these cells were more robustly activated by IL-1ß when co-cultured with astrocytes and/or BMEC. We observed a polarized endothelial response to IL-1ß, because the application of IL-1ß to the abluminal endothelial surface produced a more complex microglial inflammatory response than that which occurred following luminal IL-1ß exposure. CONCLUSIONS: Inflammatory signals are detected, amplified, and propagated through the CNS via a sequential and reverberating signaling cascade involving communication between brain endothelial cells and glia. We propose that the brain's innate immune response differs depending upon which side of the blood-brain barrier the inflammatory stimulus arises, thus allowing the brain to respond differently to central vs. peripheral inflammatory insults.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Interleukin-1beta/pharmacology , Neuroglia/metabolism , Signal Transduction/physiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/blood supply , Brain/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/drug effects , Microvessels/metabolism , Neuroglia/drug effects , Signal Transduction/drug effects
15.
PLoS One ; 11(12): e0168731, 2016.
Article in English | MEDLINE | ID: mdl-27997622

ABSTRACT

BACKGROUND: Steroid-induced sleep disturbance is a common and highly distressing morbidity for children receiving steroid chemotherapy for the treatment of pediatric acute lymphoblastic leukemia (ALL). Sleep disturbance can negatively impact overall quality of life, neurodevelopment, memory consolidation, and wound healing. Hypothalamic orexin neurons are influential wake-promoting neurons, and disturbances in orexin signaling leads to abnormal sleep behavior. A new class of drug, the orexin receptor antagonists, could be an intriguing option for sleep disorders caused by increased orexinergic output. Our aim was to examine the impact of ALL treatment doses of corticosteroids on the orexin system in rodents and in children undergoing treatment for childhood ALL. METHODS: We administered repeated injections of dexamethasone to rodents and measured responsive orexin neural activity compared to controls. In children with newly diagnosed standard risk B-cell ALL receiving dexamethasone therapy per Children's Oncology Group (COG) induction therapy from 2014-2016, we collected pre- and during-steroids matched CSF samples and measured the impact of steroids on CSF orexin concentration. RESULTS: In both rodents, all markers orexin signaling, including orexin neural output and orexin receptor expression, were preserved in the setting of dexamethasone. Additionally, we did not detect a difference in pre- and during-dexamethasone CSF orexin concentrations in children receiving dexamethasone. CONCLUSIONS: Our results demonstrate that rodent and human orexin physiology is largely preserved in the setting of high dose dexamethasone. The data obtained in our experimental model fail to demonstrate a causative role for disruption of the orexin pathway in steroid-induced sleep disturbance.


Subject(s)
Dexamethasone , Hypothalamus , Neurons/metabolism , Orexins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Signal Transduction/drug effects , Adolescent , Animals , Child , Child, Preschool , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Female , Humans , Hypothalamus/metabolism , Hypothalamus/physiopathology , Male , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Rats , Rats, Sprague-Dawley , Sleep Wake Disorders/chemically induced , Sleep Wake Disorders/metabolism , Sleep Wake Disorders/physiopathology
16.
Mol Metab ; 4(1): 25-38, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25685687

ABSTRACT

OBJECTIVE: Recent evidence indicates that the adult hematopoietic system is susceptible to diet-induced lineage skewing. It is not known whether the developing hematopoietic system is subject to metabolic programming via in utero high-fat diet (HFD) exposure, an established mechanism of adult disease in several organ systems. We previously reported substantial losses in offspring liver size with prenatal HFD. As the liver is the main hematopoietic organ in the fetus, we asked whether the developmental expansion of the hematopoietic stem and progenitor cell (HSPC) pool is compromised by prenatal HFD and/or maternal obesity. METHODS: We used quantitative assays, progenitor colony formation, flow cytometry, transplantation, and gene expression assays with a series of dietary manipulations to test the effects of gestational high-fat diet and maternal obesity on the day 14.5 fetal liver hematopoietic system. RESULTS: Maternal obesity, particularly when paired with gestational HFD, restricts physiological expansion of fetal HSPCs while promoting the opposing cell fate of differentiation. Importantly, these effects are only partially ameliorated by gestational dietary adjustments for obese dams. Competitive transplantation reveals compromised repopulation and myeloid-biased differentiation of HFD-programmed HSPCs to be a niche-dependent defect, apparent in HFD-conditioned male recipients. Fetal HSPC deficiencies coincide with perturbations in genes regulating metabolism, immune and inflammatory processes, and stress response, along with downregulation of genes critical for hematopoietic stem cell self-renewal and activation of pathways regulating cell migration. CONCLUSIONS: Our data reveal a previously unrecognized susceptibility to nutritional and metabolic developmental programming in the fetal HSPC compartment, which is a partially reversible and microenvironment-dependent defect perturbing stem and progenitor cell expansion and hematopoietic lineage commitment.

17.
PLoS One ; 9(9): e106489, 2014.
Article in English | MEDLINE | ID: mdl-25254959

ABSTRACT

Cancer cachexia is a syndrome of weight loss that results from the selective depletion of skeletal muscle mass and contributes significantly to cancer morbidity and mortality. The driver of skeletal muscle atrophy in cancer cachexia is systemic inflammation arising from both the cancer and cancer treatment. While the importance of tumor derived inflammation is well described, the mechanism by which cytotoxic chemotherapy contributes to cancer cachexia is relatively unexplored. We found that the administration of chemotherapy to mice produces a rapid inflammatory response. This drives activation of the hypothalamic-pituitary-adrenal axis, which increases the circulating level of corticosterone, the predominant endogenous glucocorticoid in rodents. Additionally, chemotherapy administration results in a significant loss of skeletal muscle mass 18 hours after administration with a concurrent induction of genes involved with the ubiquitin proteasome and autophagy lysosome systems. However, in mice lacking glucocorticoid receptor expression in skeletal muscle, chemotherapy-induced muscle atrophy is completely blocked. This demonstrates that cytotoxic chemotherapy elicits significant muscle atrophy driven by the production of endogenous glucocorticoids. Further, it argues that pharmacotherapy targeting the glucocorticoid receptor, given in concert with chemotherapy, is a viable therapeutic strategy in the treatment of cancer cachexia.


Subject(s)
Antineoplastic Agents/adverse effects , Glucocorticoids/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/pathology , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Cachexia/chemically induced , Corticosterone/blood , Female , Gene Knockout Techniques , Lysosomes/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Muscular Atrophy/blood , Muscular Atrophy/metabolism , Myocardium/metabolism , Myocardium/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Organ Size/drug effects , Organ Specificity , Proteasome Endopeptidase Complex/metabolism , Receptors, Glucocorticoid/deficiency , Receptors, Glucocorticoid/genetics , Ubiquitin/metabolism
18.
FASEB J ; 27(9): 3572-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23733748

ABSTRACT

Cachexia is a wasting condition defined by skeletal muscle atrophy in the setting of systemic inflammation. To explore the site at which inflammatory mediators act to produce atrophy in vivo, we utilized mice with a conditional deletion of the inflammatory adaptor protein myeloid differentiation factor 88 (MyD88). Although whole-body MyD88-knockout (wbMyD88KO) mice resist skeletal muscle atrophy in response to LPS, muscle-specific deletion of MyD88 is not protective. Furthermore, selective reexpression of MyD88 in the muscle of wbMyD88KO mice via electroporation fails to restore atrophy gene induction by LPS. To evaluate the role of glucocorticoids as the inflammation-induced mediator of atrophy in vivo, we generated mice with targeted deletion of the glucocorticoid receptor in muscle (mGRKO mice). Muscle-specific deletion of the glucocorticoid receptor affords a 71% protection against LPS-induced atrophy compared to control animals. Furthermore, mGRKO mice exhibit 77% less skeletal muscle atrophy than control animals in response to tumor growth. These data demonstrate that glucocorticoids are a major determinant of inflammation-induced atrophy in vivo and play a critical role in the pathogenesis of endotoxemic and cancer cachexia.


Subject(s)
Cachexia/etiology , Cachexia/metabolism , Carcinoma, Lewis Lung/physiopathology , Glucocorticoids/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Myeloid Differentiation Factor 88/metabolism , Animals , Blotting, Western , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Immunohistochemistry , In Situ Hybridization , Inflammation Mediators/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Muscular Atrophy/chemically induced , Muscular Atrophy/genetics , Myeloid Differentiation Factor 88/genetics , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
19.
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
20.
PLoS One ; 7(7): e42183, 2012.
Article in English | MEDLINE | ID: mdl-22848742

ABSTRACT

Signaling via the type 4-melanocortin receptor (MC4R) is an important determinant of body weight in mice and humans, where loss of function mutations lead to significant obesity. Humans with mutations in the MC4R experience an increase in lean mass. However, the simultaneous accrual of fat mass in such individuals may contribute to this effect via mechanical loading. We therefore examined the relationship of fat mass and lean mass in mice lacking the type-4 melanocortin receptor (MC4RKO). We demonstrate that MC4RKO mice display increased lean body mass. Further, this is not dependent on changes in adipose mass, as MC4RKO mice possess more lean body mass than diet-induced obese (DIO) wild type mice with equivalent fat mass. To examine potential sources of the increased lean mass in MC4RKO mice, bone mass and strength were examined in MC4RKO mice. Both parameters increase with age in MC4RKO mice, which likely contributes to increases in lean body mass. We functionally characterized the increased lean mass in MC4RKO mice by examining their capacity for treadmill running. MC4R deficiency results in a decrease in exercise performance. No changes in the ratio of oxidative to glycolytic fibers were seen, however MC4RKO mice demonstrate a significantly reduced heart rate, which may underlie their impaired exercise performance. The reduced exercise capacity we report in the MC4RKO mouse has potential clinical ramifications, as efforts to control body weight in humans with melanocortin deficiency may be ineffective due to poor tolerance for physical activity.


Subject(s)
Adipose Tissue/metabolism , Bone Density , Exercise Tolerance , Melanocortins/metabolism , Receptor, Melanocortin, Type 4/metabolism , Aging/metabolism , Aging/physiology , Animals , Diet/adverse effects , Female , Gene Knockout Techniques , Hand Strength/physiology , Heart Rate , Mice , Muscle Fibers, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...