Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Publication year range
1.
Cell ; 176(5): 1098-1112.e18, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30794774

ABSTRACT

Increased levels of intestinal bile acids (BAs) are a risk factor for colorectal cancer (CRC). Here, we show that the convergence of dietary factors (high-fat diet) and dysregulated WNT signaling (APC mutation) alters BA profiles to drive malignant transformations in Lgr5-expressing (Lgr5+) cancer stem cells and promote an adenoma-to-adenocarcinoma progression. Mechanistically, we show that BAs that antagonize intestinal farnesoid X receptor (FXR) function, including tauro-ß-muricholic acid (T-ßMCA) and deoxycholic acid (DCA), induce proliferation and DNA damage in Lgr5+ cells. Conversely, selective activation of intestinal FXR can restrict abnormal Lgr5+ cell growth and curtail CRC progression. This unexpected role for FXR in coordinating intestinal self-renewal with BA levels implicates FXR as a potential therapeutic target for CRC.


Subject(s)
Intestinal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/metabolism , Cell Line , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Deoxycholic Acid/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Intestinal Neoplasms/genetics , Intestines , Liver , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/physiology , Organoids/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Risk Factors , Signal Transduction , Taurocholic Acid/analogs & derivatives , Taurocholic Acid/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology
2.
Cell ; 175(1): 146-158.e15, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30100182

ABSTRACT

Pathogen virulence exists on a continuum. The strategies that drive symptomatic or asymptomatic infections remain largely unknown. We took advantage of the concept of lethal dose 50 (LD50) to ask which component of individual non-genetic variation between hosts defines whether they survive or succumb to infection. Using the enteric pathogen Citrobacter, we found no difference in pathogen burdens between healthy and symptomatic populations. Iron metabolism-related genes were induced in asymptomatic hosts compared to symptomatic or naive mice. Dietary iron conferred complete protection without influencing pathogen burdens, even at 1000× the lethal dose of Citrobacter. Dietary iron induced insulin resistance, increasing glucose levels in the intestine that were necessary and sufficient to suppress pathogen virulence. A short course of dietary iron drove the selection of attenuated Citrobacter strains that can transmit and asymptomatically colonize naive hosts, demonstrating that environmental factors and cooperative metabolic strategies can drive conversion of pathogens toward commensalism.


Subject(s)
Host-Pathogen Interactions/physiology , Iron/metabolism , Virulence/physiology , Animals , Asymptomatic Infections , Citrobacter rodentium/metabolism , Citrobacter rodentium/pathogenicity , Colitis/drug therapy , Colitis/metabolism , Colon/microbiology , Dietary Supplements , Enterobacteriaceae Infections/drug therapy , Female , Insulin Resistance/physiology , Intestine, Small/microbiology , Iron/pharmacology , Lethal Dose 50 , Male , Mice , Mice, Inbred C3H , Mice, Inbred DBA
3.
Cell ; 168(3): 503-516.e12, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28129542

ABSTRACT

Sickness-induced anorexia is a conserved behavior induced during infections. Here, we report that an intestinal pathogen, Salmonella Typhimurium, inhibits anorexia by manipulating the gut-brain axis. Inhibition of inflammasome activation by the S. Typhimurium effector, SlrP, prevented anorexia caused by IL-1ß-mediated signaling to the hypothalamus via the vagus nerve. Rather than compromising host defenses, pathogen-mediated inhibition of anorexia increased host survival. SlrP-mediated inhibition of anorexia prevented invasion and systemic infection by wild-type S. Typhimurium, reducing virulence while increasing transmission to new hosts, suggesting that there are trade-offs between transmission and virulence. These results clarify the complex and contextual role of anorexia in host-pathogen interactions and suggest that microbes have evolved mechanisms to modulate sickness-induced behaviors to promote health of their host and their transmission at the expense of virulence.


Subject(s)
Anorexia/microbiology , Salmonella Infections/microbiology , Salmonella Infections/transmission , Salmonella typhimurium/pathogenicity , Animals , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Humans , Inflammasomes/immunology , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Neural Pathways , Salmonella Infections/immunology , Salmonella typhimurium/physiology , Specific Pathogen-Free Organisms , Virulence
4.
Immunity ; 53(1): 143-157.e8, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32640256

ABSTRACT

Regulatory T (Treg) cells play a pivotal role in suppressing auto-reactive T cells and maintaining immune homeostasis. Treg cell development and function are dependent on the transcription factor Foxp3. Here, we performed a genome-wide CRISPR loss-of-function screen to identify Foxp3 regulators in mouse primary Treg cells. Foxp3 regulators were enriched in genes encoding subunits of the SWI/SNF nucleosome-remodeling and SAGA chromatin-modifying complexes. Among the three SWI/SNF-related complexes, the Brd9-containing non-canonical (nc) BAF complex promoted Foxp3 expression, whereas the PBAF complex was repressive. Chemical-induced degradation of Brd9 led to reduced Foxp3 expression and reduced Treg cell function in vitro. Brd9 ablation compromised Treg cell function in inflammatory disease and tumor immunity in vivo. Furthermore, Brd9 promoted Foxp3 binding and expression of a subset of Foxp3 target genes. Our findings provide an unbiased analysis of the genetic networks regulating Foxp3 and reveal ncBAF as a target for therapeutic manipulation of Treg cell function.


Subject(s)
CRISPR-Cas Systems/genetics , Forkhead Transcription Factors/metabolism , Neoplasms/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/metabolism , Animals , Autoimmunity/immunology , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Disease Models, Animal , Forkhead Transcription Factors/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Nucleosomes/immunology , RNA, Guide, Kinetoplastida/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics
5.
Cell ; 158(4): 734-748, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126782

ABSTRACT

The homeostasis of multicellular organisms requires terminally differentiated cells to preserve their lineage specificity. However, it is unclear whether mechanisms exist to actively protect cell identity in response to environmental cues that confer functional plasticity. Regulatory T (Treg) cells, specified by the transcription factor Foxp3, are indispensable for immune system homeostasis. Here, we report that conserved noncoding sequence 2 (CNS2), a CpG-rich Foxp3 intronic cis-element specifically demethylated in mature Tregs, helps maintain immune homeostasis and limit autoimmune disease development by protecting Treg identity in response to signals that shape mature Treg functions and drive their initial differentiation. In activated Tregs, CNS2 helps protect Foxp3 expression from destabilizing cytokine conditions by sensing TCR/NFAT activation, which facilitates the interaction between CNS2 and Foxp3 promoter. Thus, epigenetically marked cis-elements can protect cell identity by sensing key environmental cues central to both cell identity formation and functional plasticity without interfering with initial cell differentiation.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/cytology , Animals , Base Sequence , Cell Differentiation , Conserved Sequence , CpG Islands , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Mice , Molecular Sequence Data , Regulatory Sequences, Nucleic Acid , T-Lymphocytes, Regulatory/metabolism
6.
Cell ; 157(5): 1023-36, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855942

ABSTRACT

The sensation of pain is associated with increased mortality, but it is unknown whether pain perception can directly affect aging. We find that mice lacking TRPV1 pain receptors are long-lived, displaying a youthful metabolic profile at old age. Loss of TRPV1 inactivates a calcium-signaling cascade that ends in the nuclear exclusion of the CREB-regulated transcriptional coactivator CRTC1 within pain sensory neurons originating from the spinal cord. In long-lived TRPV1 knockout mice, CRTC1 nuclear exclusion decreases production of the neuropeptide CGRP from sensory endings innervating the pancreatic islets, subsequently promoting insulin secretion and metabolic health. In contrast, CGRP homeostasis is disrupted with age in wild-type mice, resulting in metabolic decline. We show that pharmacologic inactivation of CGRP receptors in old wild-type animals can restore metabolic health. These data suggest that ablation of select pain sensory receptors or the inhibition of CGRP are associated with increased metabolic health and control longevity.


Subject(s)
Longevity , Signal Transduction , TRPV Cation Channels/metabolism , Animals , CREB-Binding Protein/metabolism , Caenorhabditis elegans , Cells, Cultured , Diet , Female , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neurons/metabolism , Nociceptors/metabolism , TRPV Cation Channels/genetics , Transcription Factors/metabolism
7.
Cell ; 153(3): 601-13, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23622244

ABSTRACT

Liver fibrosis is a reversible wound-healing response involving TGFß1/SMAD activation of hepatic stellate cells (HSCs). It results from excessive deposition of extracellular matrix components and can lead to impairment of liver function. Here, we show that vitamin D receptor (VDR) ligands inhibit HSC activation by TGFß1 and abrogate liver fibrosis, whereas Vdr knockout mice spontaneously develop hepatic fibrosis. Mechanistically, we show that TGFß1 signaling causes a redistribution of genome-wide VDR-binding sites (VDR cistrome) in HSCs and facilitates VDR binding at SMAD3 profibrotic target genes via TGFß1-dependent chromatin remodeling. In the presence of VDR ligands, VDR binding to the coregulated genes reduces SMAD3 occupancy at these sites, inhibiting fibrosis. These results reveal an intersecting VDR/SMAD genomic circuit that regulates hepatic fibrogenesis and define a role for VDR as an endocrine checkpoint to modulate the wound-healing response in liver. Furthermore, the findings suggest VDR ligands as a potential therapy for liver fibrosis.


Subject(s)
Gene Regulatory Networks , Liver/metabolism , Liver/pathology , Receptors, Calcitriol/metabolism , Signal Transduction , Animals , Calcitriol/analogs & derivatives , Fibrosis/prevention & control , Genome-Wide Association Study , Hepatic Stellate Cells , Liver/injuries , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Receptors, Calcitriol/agonists , Smad3 Protein/metabolism , Transcriptome , Transforming Growth Factor beta1/metabolism
8.
Nature ; 569(7754): 131-135, 2019 05.
Article in English | MEDLINE | ID: mdl-30996350

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis largely owing to inefficient diagnosis and tenacious drug resistance. Activation of pancreatic stellate cells (PSCs) and consequent development of dense stroma are prominent features accounting for this aggressive biology1,2. The reciprocal interplay between PSCs and pancreatic cancer cells (PCCs) not only enhances tumour progression and metastasis but also sustains their own activation, facilitating a vicious cycle to exacerbate tumorigenesis and drug resistance3-7. Furthermore, PSC activation occurs very early during PDAC tumorigenesis8-10, and activated PSCs comprise a substantial fraction of the tumour mass, providing a rich source of readily detectable factors. Therefore, we hypothesized that the communication between PSCs and PCCs could be an exploitable target to develop effective strategies for PDAC therapy and diagnosis. Here, starting with a systematic proteomic investigation of secreted disease mediators and underlying molecular mechanisms, we reveal that leukaemia inhibitory factor (LIF) is a key paracrine factor from activated PSCs acting on cancer cells. Both pharmacologic LIF blockade and genetic Lifr deletion markedly slow tumour progression and augment the efficacy of chemotherapy to prolong survival of PDAC mouse models, mainly by modulating cancer cell differentiation and epithelial-mesenchymal transition status. Moreover, in both mouse models and human PDAC, aberrant production of LIF in the pancreas is restricted to pathological conditions and correlates with PDAC pathogenesis, and changes in the levels of circulating LIF correlate well with tumour response to therapy. Collectively, these findings reveal a function of LIF in PDAC tumorigenesis, and suggest its translational potential as an attractive therapeutic target and circulating marker. Our studies underscore how a better understanding of cell-cell communication within the tumour microenvironment can suggest novel strategies for cancer therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Leukemia Inhibitory Factor/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Paracrine Communication , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/diagnosis , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Humans , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/blood , Male , Mass Spectrometry , Mice , Pancreatic Neoplasms/diagnosis , Paracrine Communication/drug effects , Receptors, OSM-LIF/deficiency , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/metabolism , Tumor Microenvironment
9.
J Surg Res ; 279: 712-721, 2022 11.
Article in English | MEDLINE | ID: mdl-35933789

ABSTRACT

INTRODUCTION: Resuscitative endovascular balloon occlusion of the aorta (REBOA) causes a severe ischemia-reperfusion injury. Endovascular Perfusion Augmentation for Critical Care (EPACC) has emerged as a hemodynamic/mechanical adjunct to vasopressors and crystalloid for the treatment of post-REBOA ischemia-reperfusion injury. The objective of the study is to examine the impact of EPACC as a tool for a wean from complete REBOA compared to standard resuscitation techniques. METHODS: Nine swine underwent anesthesia and then a controlled 30% blood volume hemorrhage with 30 min of supraceliac total aortic occlusion to create an ischemia-reperfusion injury. Animals were randomized to standardized critical care (SCC) or 90 min of EPACC followed by SCC. The critical care phase lasted 270 min after injury. Hemodynamic markers and laboratory values of ischemia were recorded. RESULTS: During the first 90 min the intervention phase SCC spent 60% (54%-73%) and EPACC spent 91% (88%-92%) of the time avoiding proximal hypotension (<60 mm Hg), P = 0.03. There was also a statistically significant decrease in cumulative norepinephrine dose at the end of the experiment between SCC (80.89 mcg/kg) versus EPACC (22.03 mcg/kg), P = 0.03. Renal artery flow during EPACC was similar compared to SCC during EPACC, P = 0.19. But during the last hour of the experiment (after removal of aortic balloon) the renal artery flow in EPACC (2.9 mL/kg/min) was statistically significantly increased compared to SCC (1.57 mL/min/kg), P = 0.03. There was a statistically significant decrease in terminal creatinine in the EPACC (1.7 mg/dL) compared to SCC (2.1 mg/dL), P = 0.03. CONCLUSIONS: The 90 min of EPACC as a weaning adjunct in the setting of a severe ischemia-reperfusion injury after complete supraceliac REBOA provides improved renal flow with improvement in terminal creatinine compared to SCC with stabilized proximal hemodynamics and decreased vasopressor dose.


Subject(s)
Balloon Occlusion , Endovascular Procedures , Reperfusion Injury , Shock, Hemorrhagic , Animals , Aorta , Balloon Occlusion/methods , Creatinine , Crystalloid Solutions , Disease Models, Animal , Endovascular Procedures/methods , Norepinephrine , Perfusion , Reperfusion Injury/etiology , Reperfusion Injury/prevention & control , Resuscitation/methods , Shock, Hemorrhagic/therapy , Swine
11.
Proc Natl Acad Sci U S A ; 116(37): 18528-18536, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31455731

ABSTRACT

T helper 17 (Th17) cells produce interleukin-17 (IL-17) cytokines and drive inflammatory responses in autoimmune diseases such as multiple sclerosis. The differentiation of Th17 cells is dependent on the retinoic acid receptor-related orphan nuclear receptor RORγt. Here, we identify REV-ERBα (encoded by Nr1d1), a member of the nuclear hormone receptor family, as a transcriptional repressor that antagonizes RORγt function in Th17 cells. REV-ERBα binds to ROR response elements (RORE) in Th17 cells and inhibits the expression of RORγt-dependent genes including Il17a and Il17f Furthermore, elevated REV-ERBα expression or treatment with a synthetic REV-ERB agonist significantly delays the onset and impedes the progression of experimental autoimmune encephalomyelitis (EAE). These results suggest that modulating REV-ERBα activity may be used to manipulate Th17 cells in autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Th17 Cells/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Genetic Loci , HEK293 Cells , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Transgenic , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , RNA-Seq , Response Elements/genetics , Th17 Cells/metabolism , Thiophenes/pharmacology , Thiophenes/therapeutic use
12.
J Med Primatol ; 50(3): 164-175, 2021 06.
Article in English | MEDLINE | ID: mdl-33913156

ABSTRACT

BACKGROUND: A survey was developed to characterize disease incidence, common pathology lesions, environmental characteristics, and nutrition programs within captive research marmoset colonies. METHODS: Seventeen research facilities completed the electronic survey. RESULTS: Nutritional management programs varied amongst research institutions housing marmosets; eight primary base diets were reported. The most common clinical syndromes reported were gastrointestinal disease (i.e. inflammatory bowel disease like disease, chronic lymphocytic enteritis, chronic malabsorption, chronic diarrhea), metabolic bone disease or fracture, infectious diarrhea, and oral disease (tooth root abscesses, gingivitis, tooth root resorption). The five most common pathology morphologic diagnoses were colitis, nephropathy/nephritis, enteritis, chronic lymphoplasmacytic enteritis, and cholecystitis. Obesity was more common (average 20% of a reporting institution's population) than thin body condition (average 5%). CONCLUSIONS: Through review of current practices, we aim to inspire development of evidence-based practices to standardize husbandry and nutrition practices for marmoset research colonies.


Subject(s)
Bone Diseases, Metabolic , Callithrix , Animals , Diet/veterinary , Incidence , Obesity
13.
Nature ; 528(7580): 137-41, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26580014

ABSTRACT

Age-associated insulin resistance (IR) and obesity-associated IR are two physiologically distinct forms of adult-onset diabetes. While macrophage-driven inflammation is a core driver of obesity-associated IR, the underlying mechanisms of the obesity-independent yet highly prevalent age-associated IR are largely unexplored. Here we show, using comparative adipo-immune profiling in mice, that fat-resident regulatory T cells, termed fTreg cells, accumulate in adipose tissue as a function of age, but not obesity. Supporting the existence of two distinct mechanisms underlying IR, mice deficient in fTreg cells are protected against age-associated IR, yet remain susceptible to obesity-associated IR and metabolic disease. By contrast, selective depletion of fTreg cells via anti-ST2 antibody treatment increases adipose tissue insulin sensitivity. These findings establish that distinct immune cell populations within adipose tissue underlie ageing- and obesity-associated IR, and implicate fTreg cells as adipo-immune drivers and potential therapeutic targets in the treatment of age-associated IR.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/immunology , Aging/immunology , Insulin Resistance/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Male , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Mice , Obesity/metabolism
14.
Breast Cancer Res ; 22(1): 125, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33187540

ABSTRACT

BACKGROUND: CRIPTO is a multi-functional signaling protein that promotes stemness and oncogenesis. We previously developed a CRIPTO antagonist, ALK4L75A-Fc, and showed that it causes loss of the stem cell phenotype in normal mammary epithelia suggesting it may similarly inhibit CRIPTO-dependent plasticity in breast cancer cells. METHODS: We focused on two triple negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to measure the effects of ALK4L75A-Fc on cancer cell behavior under nutrient deprivation and endoplasmic reticulum stress. We characterized the proliferation and migration of these cells in vitro using time-lapse microscopy and characterized stress-dependent changes in the levels and distribution of CRIPTO signaling mediators and cancer stem cell markers. We also assessed the effects of ALK4L75A-Fc on proliferation, EMT, and stem cell markers in vivo as well as on tumor growth and metastasis using inducible lentiviral delivery or systemic administration of purified ALK4L75A-Fc, which represents a candidate therapeutic approach. RESULTS: ALK4L75A-Fc inhibited adaptive responses of breast cancer cells under conditions of nutrient and ER stress and reduced their proliferation, migration, clonogenicity, and expression of EMT and cancer stem cell markers. ALK4L75A-Fc also inhibited proliferation of human breast cancer cells in stressed tumor microenvironments in xenografts and reduced both primary tumor size and metastatic burden. CONCLUSIONS: Cancer cell adaptation to stresses such as nutrient deprivation, hypoxia, and chemotherapy can critically contribute to dormancy, metastasis, therapy resistance, and recurrence. Identifying mechanisms that govern cellular adaptation, plasticity, and the emergence of stem-like cancer cells may be key to effective anticancer therapies. Results presented here indicate that targeting CRIPTO with ALK4L75A-Fc may have potential as such a therapy since it inhibits breast cancer cell adaptation to microenvironmental challenges and associated stem-like and EMT phenotypes.


Subject(s)
GPI-Linked Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Recombinant Fusion Proteins/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Activin Receptors, Type I/genetics , Animals , Cell Line, Tumor , Cell Plasticity/drug effects , Endoplasmic Reticulum Stress , Female , Humans , Immunoglobulin Fc Fragments/genetics , Intercellular Signaling Peptides and Proteins , Mice , Neoplasm Recurrence, Local , Neoplastic Stem Cells/pathology , Point Mutation , Protein Binding/genetics , Protein Domains/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Triple Negative Breast Neoplasms/pathology , Tumor Hypoxia , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
15.
Proc Natl Acad Sci U S A ; 114(44): 11739-11744, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29078378

ABSTRACT

Populations of circulating immune cells are maintained in equilibrium through signals that enhance the retention or egress of hematopoietic stem cells (HSCs) from bone marrow (BM). Prostaglandin E2 (PGE2) stimulates HSC renewal and engraftment through, for example, induction of the cAMP pathway. Triggering of PGE2 receptors increases HSC survival in part via the PKA-mediated induction of the cAMP response element-binding protein (CREB) signaling pathway. PKA stimulates cellular gene expression by phosphorylating CREB at Ser133 and by promoting the dephosphorylation of the cAMP- responsive transcriptional coactivators (CRTCs). We show here that disruption of both CRTC2 and CRTC3 causes embryonic lethality, and that a single allele of either CRTC2 or CRTC3 is sufficient for viability. CRTC2 knockout mice that express one CRTC3 allele (CRTC2/3m mice) develop neutrophilia and splenomegaly in adulthood due to the up-regulation of granulocyte-colony stimulating factor (G-CSF); these effects are reversed following administration of neutralizing anti-G-CSF antiserum. Adoptive transfer of CRTC2/3m BM conferred the splenomegaly/neutrophilia phenotype in WT recipients. Targeted disruption of both CRTC2 and CRTC3 in stromal cells with a mesenchymal Prx1-Cre transgene also promoted this phenotype. Depletion of CRTC2/3 was found to decrease the expression of Suppressor of Cytokine Signaling 3 (SOCS3), leading to increases in STAT3 phosphorylation and to the induction of CEBPß, a key regulator of the G-CSF gene. As small molecule inhibition of JAK activity disrupted CEBPß induction and reduced G-CSF expression in CRTC2/3m stromal cells, our results demonstrate how cross-coupling between the CREB/CRTC and JAK/STAT pathways contributes to BM homeostasis.


Subject(s)
Bone Marrow/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Hematopoiesis/physiology , Transcription Factors/metabolism , Animals , Bone Marrow Transplantation , Embryonic Development , Gene Expression Regulation, Developmental/physiology , Granulocyte Colony-Stimulating Factor/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Transcription Factors/genetics
16.
Proc Natl Acad Sci U S A ; 114(5): 1129-1134, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096419

ABSTRACT

A fibroinflammatory stromal reaction cooperates with oncogenic signaling to influence pancreatic ductal adenocarcinoma (PDAC) initiation, progression, and therapeutic outcome, yet the mechanistic underpinning of this crosstalk remains poorly understood. Here we show that stromal cues elicit an adaptive response in the cancer cell including the rapid mobilization of a transcriptional network implicated in accelerated growth, along with anabolic changes of an altered metabolome. The close overlap of stroma-induced changes in vitro with those previously shown to be regulated by oncogenic Kras in vivo suggests that oncogenic Kras signaling-a hallmark and key driver of PDAC-is contingent on stromal inputs. Mechanistically, stroma-activated cancer cells show widespread increases in histone acetylation at transcriptionally enhanced genes, implicating the PDAC epigenome as a presumptive point of convergence between these pathways and a potential therapeutic target. Notably, inhibition of the bromodomain and extraterminal (BET) family of epigenetic readers, and of Bromodomain-containing protein 2 (BRD2) in particular, blocks stroma-inducible transcriptional regulation in vitro and tumor progression in vivo. Our work suggests the existence of a molecular "AND-gate" such that tumor activation is the consequence of mutant Kras and stromal cues, providing insight into the role of the tumor microenvironment in the origin and treatment of Ras-driven tumors.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic/genetics , Histone Code , Metabolome , Pancreatic Neoplasms/physiopathology , Stromal Cells/physiology , Tumor Microenvironment/physiology , Acetylation , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cytokines/metabolism , Energy Metabolism , Enhancer Elements, Genetic , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/physiology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Transcription Factors , Tumor Cells, Cultured
17.
Genes Dev ; 25(13): 1426-38, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21724834

ABSTRACT

Cell cycle regulation in hematopoietic stem cells (HSCs) is tightly controlled during homeostasis and in response to extrinsic stress. p53, a well-known tumor suppressor and transducer of diverse stress signals, has been implicated in maintaining HSC quiescence and self-renewal. However, the mechanisms that control its activity in HSCs, and how p53 activity contributes to HSC cell cycle control, are poorly understood. Here, we use a genetically engineered mouse to show that p53 C-terminal modification is critical for controlling HSC abundance during homeostasis and HSC and progenitor proliferation after irradiation. Preventing p53 C-terminal modification renders mice exquisitely radiosensitive due to defects in HSC/progenitor proliferation, a critical determinant for restoring hematopoiesis after irradiation. We show that fine-tuning the expression levels of the cyclin-dependent kinase inhibitor p21, a p53 target gene, contributes significantly to p53-mediated effects on the hematopoietic system. These results have implications for understanding cell competition in response to stresses involved in stem cell transplantation, recovery from adverse hematologic effects of DNA-damaging cancer therapies, and development of radioprotection strategies.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Homeostasis/genetics , Radiation Tolerance/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gamma Rays , Gene Dosage , Gene Expression Regulation , Gene Knock-In Techniques , Longevity/genetics , Male , Mice , Mice, Inbred C57BL , Mutation , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics
18.
Proc Natl Acad Sci U S A ; 112(51): 15713-8, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26644586

ABSTRACT

Liver fibrosis is characterized by the persistent deposition of extracellular matrix components by hepatic stellate cell (HSC)-derived myofibroblasts. It is the histological manifestation of progressive, but reversible wound-healing processes. An unabated fibrotic response results in chronic liver disease and cirrhosis, a pathological precursor of hepatocellular carcinoma. We report here that JQ1, a small molecule inhibitor of bromodomain-containing protein 4 (BRD4), a member of bromodomain and extraterminal (BET) proteins, abrogate cytokine-induced activation of HSCs. Cistromic analyses reveal that BRD4 is highly enriched at enhancers associated with genes involved in multiple profibrotic pathways, where BRD4 is colocalized with profibrotic transcription factors. Furthermore, we show that JQ1 is not only protective, but can reverse the fibrotic response in carbon tetrachloride-induced fibrosis in mouse models. Our results implicate that BRD4 can act as a global genomic regulator to direct the fibrotic response through its coordinated regulation of myofibroblast transcription. This suggests BRD4 as a potential therapeutic target for patients with fibrotic complications.


Subject(s)
Liver Cirrhosis, Experimental/drug therapy , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Azepines/pharmacology , Azepines/therapeutic use , Cells, Cultured , Hepatic Stellate Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Triazoles/therapeutic use
19.
Proc Natl Acad Sci U S A ; 110(17): 6991-6, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23559372

ABSTRACT

Wnt signaling in mouse mammary development and tumorigenesis has been heavily studied and characterized, but its role in human breast cancer remains elusive. Although Wnt inhibitors are in early clinical development, it is unclear whether they will be of therapeutic benefit to breast cancer patients, and subsequently, to which ones. To address this, we generated a panel of Wnt reporting human breast cancer cell lines and identified a previously unrecognized enrichment for the ability to respond to Wnt in the basal B or claudin-low subtype, which has a poor prognosis and no available targeted therapies. By co-injecting Wnt3A expressing human mammary fibroblasts with human breast cancer cell lines into mouse mammary fat pads, we showed that elevated paracrine Wnt signaling was correlated with accelerated tumor growth. Using this heterotypic system and a dual lentiviral reporter system that enables simultaneous real-time measurement of both Wnt-responsive cells and bulk tumor cells, we analyzed the outcome of elevated Wnt signaling in patient-derived xenograft (PDX) models. Interestingly, the PDX models exhibited responses not observed in the cell lines analyzed. Exogenous WNT3A promoted tumor growth in one human epidermal growth factor receptor 2-overexpressing PDX line but inhibited growth in a second PDX line obtained from a patient with triple-negative breast cancer. Tumor suppression was associated with squamous differentiation in the latter. Thus, our work suggests that paracrine Wnt signaling can either fuel or repress the growth of human breast cancers depending on yet to be determined aspects of the molecular pathways they express.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Paracrine Communication/physiology , Wnt Signaling Pathway/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Female , Fibroblasts/metabolism , Fibroblasts/transplantation , Humans , Immunohistochemistry , Luciferases , Luminescent Proteins , Mice , Mice, SCID , Receptor, ErbB-2/metabolism , Time Factors , Transplantation, Heterologous , Wnt3A Protein/metabolism , Red Fluorescent Protein
20.
Proc Natl Acad Sci U S A ; 108(8): 3412-7, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21300871

ABSTRACT

The ligand-dependent competing actions of nuclear receptor (NR)-associated transcriptional corepressor and coactivator complexes allow for the precise regulation of NR-dependent gene expression in response to both temporal and environmental cues. Here we report the mouse model termed silencing mediator of retinoid and thyroid hormone receptors (SMRT)(mRID1) in which targeted disruption of the first receptor interaction domain (RID) of the nuclear corepressor SMRT disrupts interactions with a subset of NRs and leads to diet-induced superobesity associated with a depressed respiratory exchange ratio, decreased ambulatory activity, and insulin resistance. Although apparently normal when chow fed, SMRT(mRID1) mice develop multiple metabolic dysfunctions when challenged by a high-fat diet, manifested by marked lipid accumulation in white and brown adipose tissue and the liver. The increased weight gain of SMRT(mRID1) mice on a high-fat diet occurs predominantly in fat with adipocyte hypertrophy evident in both visceral and s.c. depots. Importantly, increased inflammatory gene expression was detected only in the visceral depots. SMRT(mRID1) mice are both insulin-insensitive and refractory to the glucose-lowering effects of TZD and AICAR. Increased serum cholesterol and triglyceride levels were observed, accompanied by increased leptin and decreased adiponectin levels. Aberrant storage of lipids in the liver occurred as triglycerides and cholesterol significantly compromised hepatic function. Lipid accumulation in brown adipose tissue was associated with reduced thermogenic capacity and mitochondrial biogenesis. Collectively, these studies highlight the essential role of NR corepressors in maintaining metabolic homeostasis and describe an essential role for SMRT in regulating the progression, severity, and therapeutic outcome of metabolic diseases.


Subject(s)
Adipose Tissue/metabolism , Diet/adverse effects , Insulin Resistance , Nuclear Receptor Co-Repressor 2/physiology , Obesity/etiology , Oxidative Phosphorylation , Animals , Homeostasis , Lipid Metabolism , Liver/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL