Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Nat Commun ; 14(1): 5945, 2023 09 23.
Article En | MEDLINE | ID: mdl-37741832

Microsatellite-stable colorectal cancer (MSS-CRC) is highly refractory to immunotherapy. Understanding tumor-intrinsic determinants of immunotherapy resistance is critical to improve MSS-CRC patient outcomes. Here, we demonstrate that high tumor expression of the core autophagy gene ATG16L1 is associated with poor clinical response to anti-PD-L1 therapy in KRAS-mutant tumors from IMblaze370 (NCT02788279), a large phase III clinical trial of atezolizumab (anti-PD-L1) in advanced metastatic MSS-CRC. Deletion of Atg16l1 in engineered murine colon cancer organoids inhibits tumor growth in primary (colon) and metastatic (liver and lung) niches in syngeneic female hosts, primarily due to increased sensitivity to IFN-γ-mediated immune pressure. ATG16L1 deficiency enhances programmed cell death of colon cancer organoids induced by IFN-γ and TNF, thus increasing their sensitivity to host immunity. In parallel, ATG16L1 deficiency reduces tumor stem-like populations in vivo independently of adaptive immune pressure. This work reveals autophagy as a clinically relevant mechanism of immune evasion and tumor fitness in MSS-CRC and provides a rationale for autophagy inhibition to boost immunotherapy responses in the clinic.


Colonic Neoplasms , Colorectal Neoplasms , Animals , Female , Humans , Mice , Autophagy/genetics , Autophagy-Related Proteins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, Regulator , Liver , Clinical Trials, Phase III as Topic
2.
Cell Stem Cell ; 30(9): 1166-1178.e8, 2023 09 07.
Article En | MEDLINE | ID: mdl-37597516

The intestinal epithelium has high intrinsic turnover rate, and the precise renewal of the epithelium is dependent on the microenvironment. The intestine is innervated by a dense network of peripheral nerves that controls various aspects of intestinal physiology. However, the role of neurons in regulating epithelial cell regeneration remains largely unknown. Here, we investigated the effects of gut-innervating adrenergic nerves on epithelial cell repair following irradiation (IR)-induced injury. We observed that adrenergic nerve density in the small intestine increased post IR, while chemical adrenergic denervation impaired epithelial regeneration. Single-cell RNA sequencing experiments revealed a decrease in IL-22 signaling post IR in denervated animals. Combining pharmacologic and genetic tools, we demonstrate that ß-adrenergic receptor signaling drives IL-22 production from type 3 innate lymphoid cells (ILC3s) post IR, which in turn promotes epithelial regeneration. These results define an adrenergic-ILC3 axis important for intestinal regeneration.


Adrenergic Neurons , Immunity, Innate , Intestinal Mucosa , Lymphocytes , Regeneration , Animals , Signal Transduction , Adrenergic Neurons/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Intestinal Mucosa/physiology , Mice , Interleukin-22
3.
Mol Cell Proteomics ; 22(2): 100496, 2023 02.
Article En | MEDLINE | ID: mdl-36640924

Transcriptional enhanced associate domain family members 1 to 4 (TEADs) are a family of four transcription factors and the major transcriptional effectors of the Hippo pathway. In order to activate transcription, TEADs rely on interactions with other proteins, such as the transcriptional effectors Yes-associated protein and transcriptional co-activator with PDZ-binding motif. Nuclear protein interactions involving TEADs influence the transcriptional regulation of genes involved in cell growth, tissue homeostasis, and tumorigenesis. Clearly, protein interactions for TEADs are functionally important, but the full repertoire of TEAD interaction partners remains unknown. Here, we employed an affinity purification mass spectrometry approach to identify nuclear interacting partners of TEADs. We performed affinity purification mass spectrometry experiment in parallel in two different cell types and compared a wildtype TEAD bait protein to a nuclear localization sequence mutant that does not localize to the nucleus. We quantified the results using SAINT analysis and found a significant enrichment of proteins linked to DNA damage including X-ray repair cross-complementing protein 5 (XRCC5), X-ray repair cross-complementing protein 6 (XRCC6), poly(ADP-ribose) polymerase 1 (PARP1), and Rap1-interacting factor 1 (RIF1). In cellular assays, we found that TEADs co-localize with DNA damage-induced nuclear foci marked by histone H2AX phosphorylated on S139 (γH2AX) and Rap1-interacting factor 1. We also found that depletion of TEAD proteins makes cells more susceptible to DNA damage by various agents and that depletion of TEADs promotes genomic instability. Additionally, depleting TEADs dampens the efficiency of DNA double-stranded break repair in reporter assays. Our results connect TEADs to DNA damage response processes, positioning DNA damage as an important avenue for further research of TEAD proteins.


DNA Damage , DNA Repair , TEA Domain Transcription Factors , Humans , Carcinogenesis/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , TEA Domain Transcription Factors/metabolism
4.
Blood Adv ; 7(4): 491-507, 2023 02 28.
Article En | MEDLINE | ID: mdl-35914228

Self-renewal and differentiation of stem and progenitor cells are tightly regulated to ensure tissue homeostasis. This regulation is enabled both remotely by systemic circulating cues, such as cytokines and hormones, and locally by various niche-confined factors. R-spondin 3 (RSPO3) is one of the most potent enhancers of Wnt signaling, and its expression is usually restricted to the stem cell niche where it provides localized enhancement of Wnt signaling to regulate stem cell expansion and differentiation. Disruption of this niche-confined expression can disturb proper tissue organization and lead to cancers. Here, we investigate the consequences of disrupting the niche-restricted expression of RSPO3 in various tissues, including the hematopoietic system. We show that normal Rspo3 expression is confined to the perivascular niche in the bone marrow. Induction of increased systemic levels of circulating RSPO3 outside of the niche results in prominent loss of early B-cell progenitors and anemia but surprisingly has no effect on hematopoietic stem cells. Using molecular, pharmacologic, and genetic approaches, we show that these RSPO3-induced hematopoietic phenotypes are Wnt and RSPO3 dependent and mediated through noncanonical Wnt signaling. Our study highlights a distinct role for a Wnt/RSPO3 signaling axis in the regulation of hematopoiesis, as well as possible challenges related to therapeutic use of RSPOs for regenerative medicine.


Hematopoiesis , Stem Cell Niche , Hematopoiesis/genetics , Hematopoietic Stem Cells , Cell Differentiation/genetics , Wnt Signaling Pathway/physiology
5.
Nature ; 610(7930): 182-189, 2022 10.
Article En | MEDLINE | ID: mdl-36131013

Most current therapies that target plasma membrane receptors function by antagonizing ligand binding or enzymatic activities. However, typical mammalian proteins comprise multiple domains that execute discrete but coordinated activities. Thus, inhibition of one domain often incompletely suppresses the function of a protein. Indeed, targeted protein degradation technologies, including proteolysis-targeting chimeras1 (PROTACs), have highlighted clinically important advantages of target degradation over inhibition2. However, the generation of heterobifunctional compounds binding to two targets with high affinity is complex, particularly when oral bioavailability is required3. Here we describe the development of proteolysis-targeting antibodies (PROTABs) that tether cell-surface E3 ubiquitin ligases to transmembrane proteins, resulting in target degradation both in vitro and in vivo. Focusing on zinc- and ring finger 3 (ZNRF3), a Wnt-responsive ligase, we show that this approach can enable colorectal cancer-specific degradation. Notably, by examining a matrix of additional cell-surface E3 ubiquitin ligases and transmembrane receptors, we demonstrate that this technology is amendable for 'on-demand' degradation. Furthermore, we offer insights on the ground rules governing target degradation by engineering optimized antibody formats. In summary, this work describes a strategy for the rapid development of potent, bioavailable and tissue-selective degraders of cell-surface proteins.


Antibodies , Antibody Specificity , Membrane Proteins , Proteolysis , Ubiquitin-Protein Ligases , Animals , Antibodies/immunology , Antibodies/metabolism , Colorectal Neoplasms/metabolism , Ligands , Membrane Proteins/immunology , Membrane Proteins/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Substrate Specificity , Ubiquitin-Protein Ligases/immunology , Ubiquitin-Protein Ligases/metabolism
6.
Sci Immunol ; 6(59)2021 05 07.
Article En | MEDLINE | ID: mdl-33963061

Repair of the intestinal epithelium is tightly regulated to maintain homeostasis. The response after epithelial damage needs to be local and proportional to the insult. How different types of damage are coupled to repair remains incompletely understood. We report that after distinct types of intestinal epithelial damage, IL-1R1 signaling in GREM1+ mesenchymal cells increases production of R-spondin 3 (RSPO3), a Wnt agonist required for intestinal stem cell self-renewal. In parallel, IL-1R1 signaling regulates IL-22 production by innate lymphoid cells and promotes epithelial hyperplasia and regeneration. Although the regulation of both RSPO3 and IL-22 is critical for epithelial recovery from Citrobacter rodentium infection, IL-1R1-dependent RSPO3 production by GREM1+ mesenchymal cells alone is sufficient and required for recovery after dextran sulfate sodium-induced colitis. These data demonstrate how IL-1R1-dependent signaling orchestrates distinct repair programs tailored to the type of injury sustained that are required to restore intestinal epithelial barrier function.


Citrobacter rodentium , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/physiology , Receptors, Interleukin-1 Type I/immunology , Animals , Cells, Cultured , Coculture Techniques , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Dextran Sulfate , Epithelial Cells , Fibroblasts , Interleukins/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice, Transgenic , Organoids , Receptors, Interleukin-1 Type I/genetics , Regeneration , Signal Transduction , Thrombospondins/immunology , Interleukin-22
7.
Cell Rep Med ; 1(5): 100058, 2020 08 25.
Article En | MEDLINE | ID: mdl-33205067

The cellular origin of sporadic pancreatic neuroendocrine tumors (PNETs) is obscure. Hormone expression suggests that these tumors arise from glucagon-producing alpha cells or insulin-producing ß cells, but instability in hormone expression prevents linage determination. We utilize loss of hepatic glucagon receptor (GCGR) signaling to drive alpha cell hyperproliferation and tumor formation to identify a cell of origin and dissect mechanisms that drive progression. Using a combination of genetically engineered Gcgr knockout mice and GCGR-inhibiting antibodies, we show that elevated plasma amino acids drive the appearance of a proliferative population of SLC38A5+ embryonic progenitor-like alpha cells in mice. Further, we characterize tumors from patients with rare bi-allelic germline GCGR loss-of-function variants and find prominent tumor-cell-associated expression of the SLC38A5 paralog SLC7A8 as well as markers of active mTOR signaling. Thus, progenitor cells arise from adult alpha cells in response to metabolic signals and, when inductive signals are chronically present, drive tumor initiation.


Amino Acids/blood , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Adenoma, Islet Cell/metabolism , Adenoma, Islet Cell/pathology , Animals , Blood Glucose/metabolism , Female , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Glucagon/metabolism , Signal Transduction/physiology
8.
Methods Mol Biol ; 2171: 331-346, 2020.
Article En | MEDLINE | ID: mdl-32705654

Colorectal cancer (CRC) related death has often been attributed to the presence of metastatic disseminated disease. A concise understanding of the molecular mechanism(s) that drive metastatic progression is therefore needed but has thus far been hampered by the limited number of CRC mouse models that progress toward this disease stage. In addition, preclinical evaluation of therapeutic modalities aimed at managing metastatic disease also rests on the availability of relevant in vivo models that faithfully recapitulate the key molecular features of metastatic human CRC. To overcome these limitations, we have recently developed methodologies that enable the study of CRC progression at relevant orthotopic sites. Here, we provide a detailed methodology that describes the injection of CRC derived cell lines and organoids directly into the colorectal mucosa. This results in the growth of a single tumor mass within the colon, that can spontaneously metastasize to the liver. Furthermore, we also present a surgical procedure to directly inject cells into the portal venous circulation to induce CRC tumor growth in the liver without the requirement of a primary tumor.


Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Organoids/cytology , Animals , Disease Models, Animal , Humans , Organoids/metabolism , Xenograft Model Antitumor Assays
9.
Breast Cancer Res ; 21(1): 152, 2019 12 27.
Article En | MEDLINE | ID: mdl-31881983

BACKGROUND: PIK3CA mutations are frequent in human breast cancer. Pik3caH1047R mutant expression in mouse mammary gland promotes tumorigenesis. TP53 mutations co-occur with PIK3CA mutations in human breast cancers. We previously generated a conditionally activatable Pik3caH1047R;MMTV-Cre mouse model and found a few malignant sarcomatoid (spindle cell) carcinomas that had acquired spontaneous dominant-negative Trp53 mutations. METHODS: A Pik3caH1047R;Trp53R270H;MMTV-Cre double mutant mouse breast cancer model was generated. Tumors were characterized by histology, marker analysis, transcriptional profiling, single-cell RNA-seq, and bioinformatics. Cell lines were developed from mutant tumors and used to identify and confirm genes involved in metastasis. RESULTS: We found Pik3caH1047R and Trp53R270H cooperate in driving oncogenesis in mammary glands leading to a shorter latency than either alone. Double mutant mice develop multiple histologically distinct mammary tumors, including adenocarcinoma and sarcomatoid (spindle cell) carcinoma. We found some tumors to be invasive and a few metastasized to the lung and/or the lymph node. Single-cell RNA-seq analysis of the tumors identified epithelial, stromal, myeloid, and T cell groups. Expression analysis of the metastatic tumors identified S100a4 as a top candidate gene associated with metastasis. Metastatic tumors contained a much higher percentage of epithelial-mesenchymal transition (EMT)-signature positive and S100a4-expressing cells. CRISPR/CAS9-mediated knockout of S100a4 in a metastatic tumor-derived cell line disrupted its metastatic potential indicating a role for S100a4 in metastasis. CONCLUSIONS: Pik3caH1047R;Trp53R270H;MMTV-Cre mouse provides a preclinical model to mimic a subtype of human breast cancers that carry both PIK3CA and TP53 mutations. It also allows for understanding the cooperation between the two mutant genes in tumorigenesis. Our model also provides a system to study metastasis and develop therapeutic strategies for PIK3CA/TP53 double-positive cancers. S100a4 found involved in metastasis in this model can be a potential diagnostic and therapeutic target.


Class I Phosphatidylinositol 3-Kinases/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse , Mutation , S100 Calcium-Binding Protein A4/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Virus Infections/complications , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Cell Transformation, Viral , Class I Phosphatidylinositol 3-Kinases/genetics , Disease Models, Animal , Female , Gene Targeting , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Tumor Suppressor Protein p53/genetics , Tumor Virus Infections/virology , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 25(14): 4431-4442, 2019 07 15.
Article En | MEDLINE | ID: mdl-31004000

PURPOSE: Four consensus molecular subtypes (CMS1-4) of colorectal cancer were identified in primary tumors and found to be associated with distinctive biological features and clinical outcomes. Given that distant metastasis largely accounts for colorectal cancer-related mortality, we examined the molecular and clinical attributes of CMS in metastatic colorectal cancer (mCRC). EXPERIMENTAL DESIGN: We developed a colorectal cancer-focused NanoString-based CMS classifier that is ideally suited to interrogate archival tissues. We successfully used this panel in the CMS classification of formalin-fixed paraffin-embedded (FFPE) tissues from mCRC cohorts, one of which is composed of paired primary tumors and metastases. Finally, we developed novel mouse implantation models to enable modeling of colorectal cancer in vivo at relevant sites. RESULTS: Using our classifier, we find that the biological hallmarks of mCRC, including CMS, are in general highly similar to those observed in nonmetastatic early-stage disease. Importantly, our data demonstrate that CMS1 has the worst outcome in relapsed disease, compared with other CMS. Assigning CMS to primary tumors and their matched metastases reveals mostly concordant subtypes between primary and metastasis. Molecular analysis of matched discordant pairs reveals differences in stromal composition at each site. The development of two novel in vivo orthotopic implantation models further reinforces the notion that extrinsic factors may impact on CMS identification in matched primary and metastatic colorectal cancer. CONCLUSIONS: We describe the utility of a NanoString panel for CMS classification of FFPE clinical samples. Our work reveals the impact of intrinsic and extrinsic factors on colorectal cancer heterogeneity during disease progression.


Biomarkers, Tumor/genetics , Colorectal Neoplasms/classification , Colorectal Neoplasms/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Molecular Typing/methods , Mutation , Animals , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Cohort Studies , Colorectal Neoplasms/secondary , Female , Humans , Mice , Mice, Inbred NOD , Neoplasm Metastasis , Neoplasm Staging , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Cancer Cell ; 34(5): 792-806.e5, 2018 11 12.
Article En | MEDLINE | ID: mdl-30449325

Deregulated HER2 is a target of many approved cancer drugs. We analyzed 111,176 patient tumors and identified recurrent mutations in HER2 transmembrane domain (TMD) and juxtamembrane domain (JMD) that include G660D, R678Q, E693K, and Q709L. Using a saturation mutagenesis screen and testing of patient-derived mutations we found several activating TMD and JMD mutations. Structural modeling and analysis showed that the TMD/JMD mutations function by improving the active dimer interface or stabilizing an activating conformation. Further, we found that HER2 G660D employed asymmetric kinase dimerization for activation and signaling. Importantly, anti-HER2 antibodies and small-molecule kinase inhibitors blocked the activity of TMD/JMD mutants. Consistent with this, a G660D germline mutant lung cancer patient showed remarkable clinical response to HER2 blockade.


Lung Neoplasms/genetics , Protein Domains/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Adult , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Middle Aged , Molecular Dynamics Simulation , Mutation/genetics , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction
12.
Sci Signal ; 11(547)2018 09 11.
Article En | MEDLINE | ID: mdl-30206136

The Hippo signaling pathway regulates organ size and plays critical roles in maintaining tissue growth, homeostasis, and regeneration. Dysregulated in a wide spectrum of cancers, in mammals, this pathway is regulated by two key effectors, YAP and TAZ, that may functionally overlap. We found that TAZ promoted liver inflammation and tumor development. The expression of TAZ, but not YAP, in human liver tumors positively correlated with the expression of proinflammatory cytokines. Hyperactivated TAZ induced substantial myeloid cell infiltration into the liver and the secretion of proinflammatory cytokines through a TEAD-dependent mechanism. Furthermore, tumors with hyperactivated YAP and TAZ had distinct transcriptional signatures, which included the increased expression of inflammatory cytokines in TAZ-driven tumors. Our study elucidated a previously uncharacterized link between TAZ activity and inflammatory responses that influence tumor development in the liver.


DNA-Binding Proteins/genetics , Inflammation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Animals , Cell Cycle Proteins , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling/methods , Hippo Signaling Pathway , Humans , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred C57BL , Mutation , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , TEA Domain Transcription Factors , Trans-Activators , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transplantation, Heterologous
13.
Sci Rep ; 7(1): 12524, 2017 10 02.
Article En | MEDLINE | ID: mdl-28970566

Fibroblast Activation Protein (FAP) is a membrane-bound serine protease whose expression is often elevated in activated fibroblasts associated with tissue remodeling in various common diseases such as cancer, arthritis and fibrosis. Like the closely related dipeptidyl peptidase DPPIV, the extracellular domain of FAP can be released into circulation as a functional enzyme, and limited studies suggest that the circulating level of FAP correlates with the degree of tissue fibrosis. Here we describe a novel homogeneous fluorescence intensity assay for circulating FAP activity based on a recently identified natural substrate, FGF21. This assay is unique in that it can effectively distinguish endopeptidase activity of FAP from that of other related enzymes such as prolyl endopeptidase (PREP) and was validated using Fap-deficient mice. Structural modeling was used to elucidate the mechanistic basis for the observed specificity in substrate recognition by FAP, but not by DPPIV or PREP. Finally, the assay was used to detect elevated FAP activity in human patients diagnosed with liver cirrhosis and to determine the effectiveness of a chemical inhibitor for FAP in mice. We propose that the assay presented here could thus be utilized for diagnosis of FAP-related pathologies and for the therapeutic development of FAP inhibitors.


Fibroblast Growth Factors/genetics , Fibrosis/genetics , Gelatinases/genetics , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Serine Endopeptidases/genetics , Animals , Endopeptidases/genetics , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibrosis/enzymology , Fibrosis/pathology , Gene Expression Regulation, Enzymologic , Humans , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Mice , Prolyl Oligopeptidases , Substrate Specificity
14.
Nat Cell Biol ; 19(6): 666-676, 2017 Jun.
Article En | MEDLINE | ID: mdl-28553937

Under injury conditions, dedicated stem cell populations govern tissue regeneration. However, the molecular mechanisms that induce stem cell regeneration and enable plasticity are poorly understood. Here, we investigate stem cell recovery in the context of the hair follicle to understand how two molecularly distinct stem cell populations are integrated. Utilizing diphtheria-toxin-mediated cell ablation of Lgr5+ (leucine-rich repeat-containing G-protein-coupled receptor 5) stem cells, we show that killing of Lgr5+ cells in mice abrogates hair regeneration but this is reversible. During recovery, CD34+ (CD34 antigen) stem cells activate inflammatory response programs and start dividing. Pharmacological attenuation of inflammation inhibits CD34+ cell proliferation. Subsequently, the Wnt pathway controls the recovery of Lgr5+ cells and inhibition of Wnt signalling prevents Lgr5+ cell and hair germ recovery. Thus, our study uncovers a compensatory relationship between two stem cell populations and the underlying molecular mechanisms that enable hair follicle regeneration.


Alopecia/metabolism , Cell Plasticity , Cell Proliferation , Hair Follicle/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration , Stem Cells/metabolism , Alopecia/genetics , Alopecia/physiopathology , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, CD34/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Plasticity/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Genetic Predisposition to Disease , Hair Follicle/drug effects , Hair Follicle/growth & development , Hair Follicle/pathology , Inflammation Mediators/metabolism , Male , Mice, Knockout , Phenotype , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Regeneration/drug effects , Stem Cells/drug effects , Stem Cells/pathology , Wnt Signaling Pathway
15.
Nature ; 543(7647): 676-680, 2017 03 29.
Article En | MEDLINE | ID: mdl-28358093

Cancer stem cells (CSCs) have been hypothesized to represent the driving force behind tumour progression and metastasis, making them attractive cancer targets. However, conclusive experimental evidence for their functional relevance is still lacking for most malignancies. Here we show that the leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5) identifies intestinal CSCs in mouse tumours engineered to recapitulate the clinical progression of human colorectal cancer. We demonstrate that selective Lgr5+ cell ablation restricts primary tumour growth, but does not result in tumour regression. Instead, tumours are maintained by proliferative Lgr5- cells that continuously attempt to replenish the Lgr5+ CSC pool, leading to rapid re-initiation of tumour growth upon treatment cessation. Notably, CSCs are critical for the formation and maintenance of liver metastasis derived from colorectal cancers. Together, our data highlight distinct CSC dependencies for primary versus metastasic tumour growth, and suggest that targeting CSCs may represent a therapeutic opportunity for managing metastatic disease.


Colorectal Neoplasms/pathology , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Proliferation , Cell Separation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Disease Models, Animal , Disease Progression , Female , Injections, Subcutaneous , Intestines/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice , Neoplasm Metastasis/drug therapy , Neoplastic Stem Cells/drug effects , Organoids/pathology , Organoids/transplantation , Receptors, G-Protein-Coupled/analysis
16.
J Biol Chem ; 291(11): 5986-5996, 2016 Mar 11.
Article En | MEDLINE | ID: mdl-26797127

FGF21 is a stress-induced hormone with potent anti-obesity, insulin-sensitizing, and hepatoprotective properties. Although proteolytic cleavage of recombinant human FGF21 in preclinical species has been observed previously, the regulation of endogenously produced FGF21 is not well understood. Here we identify fibroblast activation protein (FAP) as the enzyme that cleaves and inactivates human FGF21. A selective chemical inhibitor, immunodepletion, or genetic deletion of Fap stabilized recombinant human FGF21 in serum. In addition, administration of a selective FAP inhibitor acutely increased circulating intact FGF21 levels in cynomolgus monkeys. On the basis of our findings, we propose selective FAP inhibition as a potential therapeutic approach to increase endogenous FGF21 activity for the treatment of obesity, type 2 diabetes, non-alcoholic steatohepatitis, and related metabolic disorders.


Fibroblast Growth Factors/metabolism , Gelatinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Endopeptidases , Fibroblast Growth Factors/chemistry , Gelatinases/genetics , Gene Deletion , HEK293 Cells , Humans , Macaca fascicularis , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine Endopeptidases/genetics
17.
Nature ; 529(7584): 97-100, 2016 Jan 07.
Article En | MEDLINE | ID: mdl-26700806

Colorectal cancer remains a major unmet medical need, prompting large-scale genomics efforts in the field to identify molecular drivers for which targeted therapies might be developed. We previously reported the identification of recurrent translocations in R-spondin genes present in a subset of colorectal tumours. Here we show that targeting RSPO3 in PTPRK-RSPO3-fusion-positive human tumour xenografts inhibits tumour growth and promotes differentiation. Notably, genes expressed in the stem-cell compartment of the intestine were among those most sensitive to anti-RSPO3 treatment. This observation, combined with functional assays, suggests that a stem-cell compartment drives PTPRK-RSPO3 colorectal tumour growth and indicates that the therapeutic targeting of stem-cell properties within tumours may be a clinically relevant approach for the treatment of colorectal tumours.


Cell Differentiation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Thrombospondins/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibodies/therapeutic use , Cell Division/drug effects , Colorectal Neoplasms/metabolism , Disease Progression , Female , Gene Expression Regulation/drug effects , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/drug effects , Intestines/pathology , Male , Mice , Neoplastic Stem Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Thrombospondins/antagonists & inhibitors , Thrombospondins/immunology , Xenograft Model Antitumor Assays
18.
Cell Rep ; 12(3): 495-510, 2015 Jul 21.
Article En | MEDLINE | ID: mdl-26166562

Understanding the regulation of islet cell mass has important implications for the discovery of regenerative therapies for diabetes. The liver plays a central role in metabolism and the regulation of endocrine cell number, but liver-derived factors that regulate α-cell and ß-cell mass remain unidentified. We propose a nutrient-sensing circuit between liver and pancreas in which glucagon-dependent control of hepatic amino acid metabolism regulates α-cell mass. We found that glucagon receptor inhibition reduced hepatic amino acid catabolism, increased serum amino acids, and induced α-cell proliferation in an mTOR-dependent manner. In addition, mTOR inhibition blocked amino-acid-dependent α-cell replication ex vivo and enabled conversion of α-cells into ß-like cells in vivo. Serum amino acids and α-cell proliferation were increased in neonatal mice but fell throughout postnatal development in a glucagon-dependent manner. These data reveal that amino acids act as sensors of glucagon signaling and can function as growth factors that increase α-cell proliferation.


Amino Acids/metabolism , Glucagon/metabolism , Liver/cytology , Liver/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Proliferation , Metabolism , Mice , Signal Transduction
19.
Genes Cancer ; 6(3-4): 106-18, 2015 Mar.
Article En | MEDLINE | ID: mdl-26000094

Lung adenocarcinoma is often discovered as metastatic disease with very poor prognosis. However, much remains unknown about the mechanisms of lung adenocarcinoma tumor progression. In this study we showed that knockdown of BUB1B/BUBR1, a critical mitotic checkpoint protein, significantly inhibited anchorage-independent growth of lung adenocarcinoma cell lines. In allograft and tail vein mouse model studies, BUB1B suppression inhibited primary tumor growth and reduced metastasis to the lung and lymph nodes, resulting in prolonged survival in both tumor prevention and tumor intervention settings. Mechanistic studies revealed that BUB1B knockdown sensitized cells to anoikis. The N-terminal region and GLEBS domain of BUB1B were required for its functions in both anchorage-independent growth and anoikis resistance, whereas the kinase domain was less critical. Overexpression of BUB1B is associated with disease progression and poor survival in human lung adenocarcinoma patients. Collectively, these data reveal a novel function for BUB1B in mediating anchorage-independent survival and growth, thereby facilitating lung adenocarcinoma dissemination during metastasis. Thus, targeting BUB1B could provide potential therapeutic benefit in suppressing metastasis and prolonging survival in lung adenocarcinoma patients.

20.
Proc Natl Acad Sci U S A ; 111(38): 13942-7, 2014 Sep 23.
Article En | MEDLINE | ID: mdl-25201978

Mammalian hosts are colonized with commensal microbes in various mucosal and epithelial tissues, including the intestinal tract. In mice, the presence of segmented filamentous bacteria (SFB) promotes Th17 differentiation and the development of autoimmune disease. Here, we demonstrate that the IL-23 pathway dynamically regulates the abundance of SFB as well as mucosal barrier function in the adult animal. Genetic or pharmacological inactivation of the pathway selectively perturbs the abundance of a small group of commensals, including SFB, and results in an impaired mucosal barrier. Defective barrier function leads to systemic dissemination of microbial products, provoking induction of the IL-23 pathway with dual consequences: IL-23 drives IL-22 production to reinforce mucosal barrier function and elicit antimicrobial activities, and it also drives the differentiation of Th17 cells in an attempt to combat escaped microbes in the lamina propria and in distal tissues. Thus, barrier defects generate a systemic environment that facilitates Th17 development.


Interleukins/immunology , Intestinal Mucosa/immunology , Microbiota/immunology , Receptors, Interleukin/immunology , Th17 Cells/immunology , Animals , Cell Differentiation/immunology , Interleukins/genetics , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Receptors, Interleukin/genetics , Interleukin-22
...