Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Nat Commun ; 14(1): 8075, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38092754

ABSTRACT

The metabolic and signaling pathways regulating aggressive mesenchymal colorectal cancer (CRC) initiation and progression through the serrated route are largely unknown. Although relatively well characterized as BRAF mutant cancers, their poor response to current targeted therapy, difficult preneoplastic detection, and challenging endoscopic resection make the identification of their metabolic requirements a priority. Here, we demonstrate that the phosphorylation of SCAP by the atypical PKC (aPKC), PKCλ/ι promotes its degradation and inhibits the processing and activation of SREBP2, the master regulator of cholesterol biosynthesis. We show that the upregulation of SREBP2 and cholesterol by reduced aPKC levels is essential for controlling metaplasia and generating the most aggressive cell subpopulation in serrated tumors in mice and humans. Since these alterations are also detected prior to neoplastic transformation, together with the sensitivity of these tumors to cholesterol metabolism inhibitors, our data indicate that targeting cholesterol biosynthesis is a potential mechanism for serrated chemoprevention.


Subject(s)
Protein Kinase C , Signal Transduction , Animals , Humans , Mice , Cell Transformation, Neoplastic/genetics , Cholesterol , Epithelial Cells/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism
2.
Nat Commun ; 14(1): 5534, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37749092

ABSTRACT

Mesenchymal activation, characterized by dense stromal infiltration of immune and mesenchymal cells, fuels the aggressiveness of colorectal cancers (CRC), driving progression and metastasis. Targetable molecules in the tumor microenvironment (TME) need to be identified to improve the outcome in CRC patients with this aggressive phenotype. This study reports a positive link between high thrombospondin-1 (THBS1) expression and mesenchymal characteristics, immunosuppression, and unfavorable CRC prognosis. Bone marrow-derived monocyte-like cells recruited by CXCL12 are the primary source of THBS1, which contributes to the development of metastasis by inducing cytotoxic T-cell exhaustion and impairing vascularization. Furthermore, in orthotopically generated CRC models in male mice, THBS1 loss in the TME renders tumors partially sensitive to immune checkpoint inhibitors and anti-cancer drugs. Our study establishes THBS1 as a potential biomarker for identifying mesenchymal CRC and as a critical suppressor of antitumor immunity that contributes to the progression of this malignancy with a poor prognosis.


Subject(s)
Colorectal Neoplasms , Monocytes , Humans , Male , Animals , Mice , Immunosuppression Therapy , Aggression , Immune Checkpoint Inhibitors , Tumor Microenvironment
3.
STAR Protoc ; 4(2): 102243, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37083323

ABSTRACT

Imaging organoid culture provides an excellent tool for studying complex diseases such as cancer. However, retaining the morphology of intact organoids for immunolabeling has been challenging. Here, we describe a protocol for immunofluorescence staining in intact colorectal cancer organoids derived from mice. We also describe additional steps for co-culture with mouse fibroblasts to enable the study of interactions with other cellular components of the tissue microenvironment. For complete details on the use and execution of this protocol, please refer to Martinez-Ordoñez et al. (2023).1.

4.
Cancer Cell ; 41(2): 252-271.e9, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36525970

ABSTRACT

Mesenchymal colorectal cancer (mCRC) is microsatellite stable (MSS), highly desmoplastic, with CD8+ T cells excluded to the stromal periphery, resistant to immunotherapy, and driven by low levels of the atypical protein kinase Cs (aPKCs) in the intestinal epithelium. We show here that a salient feature of these tumors is the accumulation of hyaluronan (HA) which, along with reduced aPKC levels, predicts poor survival. HA promotes epithelial heterogeneity and the emergence of a tumor fetal metaplastic cell (TFMC) population endowed with invasive cancer features through a network of interactions with activated fibroblasts. TFMCs are sensitive to HA deposition, and their metaplastic markers have prognostic value. We demonstrate that in vivo HA degradation with a clinical dose of hyaluronidase impairs mCRC tumorigenesis and liver metastasis and enables immune checkpoint blockade therapy by promoting the recruitment of B and CD8+ T cells, including a proportion with resident memory features, and by blocking immunosuppression.


Subject(s)
Colorectal Neoplasms , Hyaluronic Acid , Tumor Microenvironment , Humans , CD8-Positive T-Lymphocytes/pathology , Colorectal Neoplasms/pathology , Hyaluronic Acid/metabolism , Immunotherapy , Sarcoma/pathology , Tumor Microenvironment/physiology
5.
STAR Protoc ; 3(3): 101635, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36035805

ABSTRACT

Air-liquid organotypic culture models enable the study of the cellular crosstalk in the tumor microenvironment. This 3D assay recapitulates the tumor niche more faithfully than 2D culture systems and represents a versatile platform that can be easily adapted to different types of cancer cells, stromal components, or ECM composition. Here, we detail the steps to build an organotypic culture including the preparation of the organotypic structure, organotypic gels, cell seeding, gel casting, membrane processing, and image and data analysis. For complete details on the use and execution of this protocol, please refer to Linares et al. (2022).


Subject(s)
Neoplasms , Tumor Microenvironment , Cell Line, Tumor
6.
Cell Rep ; 39(6): 110792, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545049

ABSTRACT

Reduced p62 levels are associated with the induction of the cancer-associated fibroblast (CAF) phenotype, which promotes tumorigenesis in vitro and in vivo through inflammation and metabolic reprogramming. However, how p62 is downregulated in the stroma fibroblasts by tumor cells to drive CAF activation is an unresolved central issue in the field. Here we show that tumor-secreted lactate downregulates p62 transcriptionally through a mechanism involving reduction of the NAD+/NADH ratio, which impairs poly(ADP-ribose)-polymerase 1 (PARP-1) activity. PARP-1 inhibition blocks the poly(ADP-ribosyl)ation of the AP-1 transcription factors, c-FOS and c-JUN, which is an obligate step for p62 downregulation. Importantly, restoring p62 levels in CAFs by NAD+ renders CAFs less active. PARP inhibitors, such as olaparib, mimick lactate in the reduction of stromal p62 levels, as well as the subsequent stromal activation both in vitro and in vivo, which suggests that therapies using olaparib would benefit from strategies aimed at inhibiting CAF activity.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Cancer-Associated Fibroblasts/metabolism , Fibroblasts/metabolism , Lactic Acid/metabolism , NAD/metabolism , Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism
7.
Trends Cell Biol ; 32(12): 1023-1034, 2022 12.
Article in English | MEDLINE | ID: mdl-35501226

ABSTRACT

Nononcogenic cancer drivers often impinge on complex signals that create new addictions and vulnerabilities. Protein kinase Cλ/ι (PKCλ/ι) suppresses tumorigenesis by blocking metabolic pathways that regulate fuel oxidation and create building blocks for the epigenetic control of cell differentiation. Reduced levels of PKCλ/ι unleash these pathways to promote tumorigenesis, but the simultaneous activation of the STING-driven interferon cascade prevents tumor initiation by triggering immunosurveillance mechanisms. However, depending on the context of other signaling pathways, such as WNT/ß-catenin or PKCζ, and timing, PKCλ/ι deletion can promote or inhibit tumorigenesis. In this review, we discuss in detail the molecular and cellular underpinnings of PKCλ/ι functions in cancer with the perspective of the crosstalk between metabolism and inflammation in the tumor microenvironment.


Subject(s)
Isoenzymes , Neoplasms , Humans , Isoenzymes/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction/physiology , Neoplasms/genetics , Cell Transformation, Neoplastic/pathology , Tumor Microenvironment
8.
Mol Cell ; 81(21): 4509-4526.e10, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34560002

ABSTRACT

The interferon (IFN) pathway is critical for cytotoxic T cell activation, which is central to tumor immunosurveillance and successful immunotherapy. We demonstrate here that PKCλ/ι inactivation results in the hyper-stimulation of the IFN cascade and the enhanced recruitment of CD8+ T cells that impaired the growth of intestinal tumors. PKCλ/ι directly phosphorylates and represses the activity of ULK2, promoting its degradation through an endosomal microautophagy-driven ubiquitin-dependent mechanism. Loss of PKCλ/ι results in increased levels of enzymatically active ULK2, which, by direct phosphorylation, activates TBK1 to foster the activation of the STING-mediated IFN response. PKCλ/ι inactivation also triggers autophagy, which prevents STING degradation by chaperone-mediated autophagy. Thus, PKCλ/ι is a hub regulating the IFN pathway and three autophagic mechanisms that serve to maintain its homeostatic control. Importantly, single-cell multiplex imaging and bioinformatics analysis demonstrated that low PKCλ/ι levels correlate with enhanced IFN signaling and good prognosis in colorectal cancer patients.


Subject(s)
Colorectal Neoplasms/metabolism , Interferons/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/physiology , Signal Transduction , Adult , Aged , Aged, 80 and over , Animals , Autophagy , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis , Cell Transformation, Neoplastic , Colorectal Neoplasms/mortality , Cycloheximide/chemistry , Female , HEK293 Cells , Humans , Immunophenotyping , Interferon Regulatory Factor-3/metabolism , Male , Membrane Proteins/metabolism , Mice , Middle Aged , Neoplasm Transplantation , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , Up-Regulation
9.
Nat Commun ; 12(1): 2876, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001883

ABSTRACT

Activation of non-shivering thermogenesis is considered a promising approach to lower body weight in obesity. p62 deficiency in adipocytes reduces systemic energy expenditure but its role in sustaining mitochondrial function and thermogenesis remains unresolved. NBR1 shares a remarkable structural similarity with p62 and can interact with p62 through their respective PB1 domains. However, the physiological relevance of NBR1 in metabolism, as compared to that of p62, was not clear. Here we show that whole-body and adipocyte-specific ablation of NBR1 reverts the obesity phenotype induced by p62 deficiency by restoring global energy expenditure and thermogenesis in brown adipose tissue. Impaired adrenergic-induced browning of p62-deficient adipocytes is rescued by NBR1 inactivation, unveiling a negative role of NBR1 in thermogenesis under conditions of p62 loss. We demonstrate that upon p62 inactivation, NBR1 represses the activity of PPARγ, establishing an unexplored p62/NBR1-mediated paradigm in adipocyte thermogenesis that is critical for the control of obesity.


Subject(s)
Adipocytes/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , PPAR gamma/metabolism , Sequestosome-1 Protein/deficiency , Thermogenesis , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Animals, Newborn , Cell Nucleus/metabolism , Cells, Cultured , Energy Metabolism/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PPAR gamma/genetics , Protein Binding , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Sequestosome-1 Protein/genetics
10.
STAR Protoc ; 2(1): 100297, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33554135

ABSTRACT

In vivo interrogation of the functional role of genes implicated in colorectal cancer (CRC) is limited by the need for physiological models that mimic the disease. Here, we describe a protocol that provides the steps required for the orthotopic co-implantation of tumoral and stromal cells into the cecum and rectum to investigate the crosstalk between the tumor and its microenvironment. This protocol recapitulates metastases to the lymph nodes, liver, and lungs observed in human CRC. For complete details on the use and execution of this protocol, please refer to Kasashima et al. (2020).


Subject(s)
Cecum/metabolism , Colorectal Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Rectum/metabolism , Tumor Microenvironment , Animals , Cecum/pathology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Mice , Neoplasms, Experimental/pathology , Rectum/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
11.
Dev Cell ; 56(1): 95-110.e10, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33207226

ABSTRACT

Cancer-associated fibroblasts (CAFs) promote tumor malignancy, but the precise transcriptional mechanisms regulating the acquisition of the CAF phenotype are not well understood. We show that the upregulation of SOX2 is central to this process, which is repressed by protein kinase Cζ (PKCζ). PKCζ deficiency activates the reprogramming of colonic fibroblasts to generate a predominant SOX2-dependent CAF population expressing the WNT regulator Sfrp2 as its top biomarker. SOX2 directly binds the Sfrp1/2 promoters, and the inactivation of Sox2 or Sfrp1/2 in CAFs impaired the induction of migration and invasion of colon cancer cells, as well as their tumorigenicity in vivo. Importantly, recurrence-free and overall survival of colorectal cancer (CRC) patients negatively correlates with stromal PKCζ levels. Also, SOX2 expression in the stroma is associated with CRC T invasion and worse prognosis of recurrence-free survival. Therefore, the PKCζ-SOX2 axis emerges as a critical step in the control of CAF pro-tumorigenic potential.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinogenesis/metabolism , Colorectal Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Kinase C/deficiency , SOXB1 Transcription Factors/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Invasiveness/genetics , Organoids/metabolism , Organoids/pathology , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA-Seq , Recurrence , SOXB1 Transcription Factors/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Single-Cell Analysis , Up-Regulation , beta Catenin/genetics , beta Catenin/metabolism
12.
STAR Protoc ; 1(3): 100185, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377079

ABSTRACT

This protocol provides the steps required for a mouse liver orthotopic implantation model. The reliable pre-clinical animal models that have similar characteristics to hepatocellular carcinoma (HCC) are a powerful tool to unveil the mechanisms controlling tumor initiation and progression. Here, we describe a syngeneic orthotopic HCC model that recapitulates the role of a host pro-tumorigenic microenvironment by pre-conditioning mouse livers with a high-fat diet (HFD). For complete details on the use and execution of this protocol, please refer to Kudo et al. (2020).


Subject(s)
Carcinoma, Hepatocellular/pathology , Fatty Liver/complications , Liver Neoplasms/pathology , Animals , Disease Models, Animal , Liver/pathology , Mice, Inbred C57BL , Sutures
13.
Cancer Cell ; 38(2): 247-262.e11, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32589943

ABSTRACT

Oxidative stress plays a critical role in liver tissue damage and in hepatocellular carcinoma (HCC) initiation and progression. However, the mechanisms that regulate autophagy and metabolic reprogramming during reactive oxygen species (ROS) generation, and how ROS promote tumorigenesis, still need to be fully understood. We show that protein kinase C (PKC) λ/ι loss in hepatocytes promotes autophagy and oxidative phosphorylation. This results in ROS generation, which through NRF2 drives HCC through cell-autonomous and non-autonomous mechanisms. Although PKCλ/ι promotes tumorigenesis in oncogene-driven cancer models, emerging evidence demonstrate that it is a tumor suppressor in more complex carcinogenic processes. Consistently, PKCλ/ι levels negatively correlate with HCC histological tumor grade, establishing this kinase as a tumor suppressor in liver cancer.


Subject(s)
Autophagy/genetics , Carcinoma, Hepatocellular/genetics , Isoenzymes/genetics , Liver Neoplasms/genetics , NF-E2-Related Factor 2/genetics , Oxidative Phosphorylation , Protein Kinase C/genetics , RNA Interference , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Disease Progression , HEK293 Cells , Hep G2 Cells , Humans , Isoenzymes/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Protein Kinase C/metabolism
14.
Rev. clín. med. fam ; 13(2): 131-138, jun. 2020. tab, graf
Article in Spanish | IBECS | ID: ibc-199834

ABSTRACT

OBJETIVO: Determinar los patrones de susceptibilidad antimicrobiana de bacterias Gram negativas aisladas de cultivos de orina de pacientes ambulatorios, y asociarlos con variables como edad, sexo, infección urinaria previa y presencia de diabetes mellitus tipo 2. MATERIAL Y MÉTODOS: Estudio descriptivo observacional y trasversal, con cepas aisladas de 278 pacientes con infección urinaria baja, que acudieron a consulta externa a la CMF Dr. Ignacio Chávez del Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, del sur de la Ciudad de México, entre los meses de marzo de 2018 a febrero de 2019. Se utilizó el sistema Phoenix 100 de Becton Dickinson, tanto para identificación de cepas, como para determinación de susceptibilidad antimicrobiana. Se probaron 16 antimicrobianos. Se utilizó estadística descriptiva para determinar proporción de resistencias. Programa estadístico SPSS versión 22. RESULTADOS: Se incluyeron 278 cepas: 231 Escherichia coli, 24 Klebsiella spp; 8 Enterobacter spp, 7 Proteus spp, 7 Citrobacter spp, 1 Serratia spp. La mayor resistencia fue para: ampicilina con 74,1 %, y la mayor sensibilidad para amikacina con 100 %. Del total de cepas, 140 (50,4 %) fueron Multi-Drogo-Resistentes, no hubo cepas Pan-Drogo-Resistentes. Al asociar las variables de estudio con la resistencia a cada antimicrobiano, sólo se obtuvo significancia estadística entre las cefalosporinas cefaxolina y cefoxitin, y el imipenem con el sexo de los pacientes, con mayor porcentaje en los hombres. CONCLUSIONES: Se obtuvo una alta resistencia en prácticamente todos los grupos de antimicrobianos, lo que hace necesario estar al tanto de los patrones de susceptibilidad en cada zona o país


OBJECTIVES: To determine patterns of antimicrobial susceptibility of Gram-negative bacteria isolated from urine cultures of outpatients, and to associate them with variables such as age, sex, previous urinary infection, and presence of type-2 diabetes mellitus. MATERIAL AND METHODS: Descriptive, observational, cross-sectional study, with strains isolated from 278 patients with low urinary infection, who had outpatient consultation in CMF Dr. Ignacio Chávez of the Instituto de Seguridad y Servicios Sociales de los Trabajadoresdel Estado, in southern Ciudad de México, between March 2018 and February 2019. The Becton-Dickinson Phoenix 100 system was used both for identifying strains, and for determining antimicrobial susceptibility. 16 antimicrobial agents were tested. Descriptive statistics was used for determining resistance proportion. Statistical programme SPSS version 22. RESULTS: 278 strains were included: 231 Escherichia coli, 24 Klebsiella spp., 8 Enterobacter spp., 7 Proteus spp., 7 Citrobacter spp., 1 Serratia spp. The greatest resistance was for: ampicillin with 74.1% and the greatest sensitivity was for amikacin with 100%. Of all strains, 140 (50.4%) were multidrug resistant, and none was pandrug resistant. When the study variables were associated with resistance to each antimicrobial, there was statistical significance only between cephalosporins cefazolin and cefoxitin, as well as imipenem with patient sex, with higher percentage in men. CONCLUSIONS: High resistance was obtained in virtually all the groups of antimicrobials. This makes it necessary to be aware of susceptibility patterns in each area or country


Subject(s)
Humans , Male , Female , Child, Preschool , Child , Adolescent , Young Adult , Adult , Middle Aged , Aged , Aged, 80 and over , Microbial Sensitivity Tests/methods , Anti-Bacterial Agents/pharmacokinetics , Gram-Negative Bacterial Infections/drug therapy , Urinary Tract Infections/drug therapy , Escherichia coli Infections/drug therapy , Drug Resistance, Microbial , In Vitro Techniques/methods , Mexico/epidemiology , Gram-Negative Bacteria/pathogenicity , Escherichia coli/pathogenicity , Diabetes Mellitus, Type 2/complications , Ambulatory Care/statistics & numerical data , Cross-Sectional Studies/statistics & numerical data
15.
J Hepatol ; 72(6): 1182-1195, 2020 06.
Article in English | MEDLINE | ID: mdl-32105670

ABSTRACT

BACKGROUND & AIMS: Hepatomegaly can be triggered by insulin and insulin-unrelated etiologies. Insulin acts via AKT, but how other challenges cause hepatomegaly is unknown. METHODS: Since many hepatomegaly-inducing toxicants and stressors activate NRF2, we examined the effect of NRF2 activation on liver size and metabolism using a conditional allele encoding a constitutively active NRF2 variant to generate Nrf2Act-hep mice in which NRF2 is selectively activated in hepatocytes. We also used adenoviruses encoding variants of the autophagy adaptor p62/SQSTM1, which activates liver NRF2, as well as liver-specific ATG7-deficient mice (Atg7Δhep) and liver specimens from patients with hepatic sinusoidal obstruction syndrome (HSOS) and autoimmune hepatitis (AIH). RNA sequencing and cell signaling analyses were used to determine cellular consequences of NRF2 activation and diverse histological analyses were used to study effects of the different manipulations on liver and systemic pathophysiology. RESULTS: Hepatocyte-specific NRF2 activation, due to p62 accumulation or inhibition of KEAP1 binding, led to hepatomegaly associated with enhanced glycogenosis, steatosis and G2/M cell cycle arrest, fostering hyperplasia without cell division. Surprisingly, all manipulations that led to NRF2 activation also activated AKT, whose inhibition blocked NRF2-induced hepatomegaly and glycogenosis, but not NRF2-dependent antioxidant gene induction. AKT activation was linked to NRF2-mediated transcriptional induction of PDGF and EGF receptor ligands that signaled through their cognate receptors in an autocrine manner. Insulin and insulin-like growth factors were not involved. The NRF2-AKT signaling axis was also activated in human HSOS- and AIH-related hepatomegaly. CONCLUSIONS: NRF2, a transcription factor readily activated by xenobiotics, oxidative stress and autophagy disruptors, may be a common mediator of hepatomegaly; its effects on hepatic metabolism can be reversed by AKT/tyrosine kinase inhibitors. LAY SUMMARY: Hepatomegaly can be triggered by numerous etiological factors, including infections, liver cancer, metabolic disturbances, toxicant exposure, as well as alcohol abuse or drug-induced hepatitis. This study identified the oxidative stress response transcription factor NRF2 as a common mediator of hepatomegaly. NRF2 activation results in elevated expression of several growth factors. These growth factors are made by hepatocytes and activate their receptors in an autocrine fashion to stimulate the accumulation of glycogen and lipids that lead to hepatocyte and liver enlargement. The protein kinase AKT plays a key role in this process and its inhibition leads to reversal of hepatomegaly.


Subject(s)
ErbB Receptors/metabolism , Genes, erbB-1 , Hepatic Veno-Occlusive Disease/complications , Hepatic Veno-Occlusive Disease/metabolism , Hepatitis, Autoimmune/complications , Hepatitis, Autoimmune/metabolism , Hepatomegaly/complications , Hepatomegaly/metabolism , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adult , Animals , Autophagy/genetics , Disease Models, Animal , ErbB Receptors/genetics , Female , Hemangioma/metabolism , Hemangioma/pathology , Hepatic Veno-Occlusive Disease/pathology , Hepatitis, Autoimmune/pathology , Hepatomegaly/genetics , Hepatomegaly/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/genetics
16.
Cancer Cell ; 35(3): 385-400.e9, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30827887

ABSTRACT

Increasingly effective therapies targeting the androgen receptor have paradoxically promoted the incidence of neuroendocrine prostate cancer (NEPC), the most lethal subtype of castration-resistant prostate cancer (PCa), for which there is no effective therapy. Here we report that protein kinase C (PKC)λ/ι is downregulated in de novo and during therapy-induced NEPC, which results in the upregulation of serine biosynthesis through an mTORC1/ATF4-driven pathway. This metabolic reprogramming supports cell proliferation and increases intracellular S-adenosyl methionine (SAM) levels to feed epigenetic changes that favor the development of NEPC characteristics. Altogether, we have uncovered a metabolic vulnerability triggered by PKCλ/ι deficiency in NEPC, which offers potentially actionable targets to prevent therapy resistance in PCa.


Subject(s)
Carcinoma, Neuroendocrine/pathology , Down-Regulation , Isoenzymes/deficiency , Prostatic Neoplasms/pathology , Protein Kinase C/deficiency , Serine/metabolism , Activating Transcription Factor 4/metabolism , Biosynthetic Pathways , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Cell Line, Tumor , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , S-Adenosylmethionine/metabolism
17.
Immunity ; 49(6): 1132-1147.e7, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30552022

ABSTRACT

Serrated adenocarcinoma, an alternative pathway for colorectal cancer (CRC) development, accounts for 15%-30% of all CRCs and is aggressive and treatment resistant. We show that the expression of atypical protein kinase C ζ (PKCζ) and PKCλ/ι was reduced in human serrated tumors. Simultaneous inactivation of the encoding genes in the mouse intestinal epithelium resulted in spontaneous serrated tumorigenesis that progressed to advanced cancer with a strongly reactive and immunosuppressive stroma. Whereas epithelial PKCλ/ι deficiency led to immunogenic cell death and the infiltration of CD8+ T cells, which repressed tumor initiation, PKCζ loss impaired interferon and CD8+ T cell responses, which resulted in tumorigenesis. Combined treatment with a TGF-ß receptor inhibitor plus anti-PD-L1 checkpoint blockade showed synergistic curative activity. Analysis of human samples supported the relevance of these kinases in the immunosurveillance defects of human serrated CRC. These findings provide insight into avenues for the detection and treatment of this poor-prognosis subtype of CRC.


Subject(s)
Intestinal Mucosa/immunology , Intestinal Neoplasms/immunology , Isoenzymes/immunology , Protein Kinase C/immunology , Adult , Aged , Aged, 80 and over , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Female , Humans , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice, Knockout , Mice, Transgenic , Middle Aged , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism
18.
Cell Rep ; 23(4): 1178-1191, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29694894

ABSTRACT

Most colorectal cancer (CRC)-related deaths are due to liver metastases. PKCζ is a tumor suppressor in CRC with reduced expression in metastasis. Given the importance of microRNAs (miRNAs) in regulating cellular plasticity, we performed an unbiased screening and identified the miR-200 family as the most relevant miRNAs downregulated by PKCζ deficiency. The regulation of the intracellular levels of miR-200 by PKCζ is post-transcriptional and involves their secretion in extracellular vesicles. Here, we identified ADAR2 as a direct substrate of PKCζ in CRC cells. Phosphorylation of ADAR2 regulates its editing activity, which is required to maintain miR-200 steady-state levels, suggesting that the PKCζ/ADAR2 axis regulates miR-200 secretion through RNA editing. Loss of this axis results in epithelial-to-mesenchymal transition (EMT) and increased liver metastases, which can be inhibited in vivo by blocking miR-200 release. Therefore, the PKCζ/ADAR2 axis is a critical regulator of CRC metastases through modulation of miR-200 levels.


Subject(s)
Adenosine Deaminase , Circulating MicroRNA , Colorectal Neoplasms , Liver Neoplasms , MicroRNAs , Neoplasm Proteins , Protein Kinase C , RNA, Neoplasm , RNA-Binding Proteins , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Cell-Derived Microparticles/genetics , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Circulating MicroRNA/genetics , Circulating MicroRNA/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
19.
Cancer Cell ; 33(4): 770-784.e6, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29634950

ABSTRACT

Obesity is a leading risk factor for cancer. However, understanding the crosstalk between adipocytes and tumor cells in vivo, independently of dietary contributions, is a major gap in the field. Here we used a prostate cancer (PCa) mouse model in which the signaling adaptor p62/Sqstm1 is selectively inactivated in adipocytes. p62 loss in adipocytes results in increased osteopontin secretion, which mediates tumor fatty acid oxidation and invasion, leading to aggressive metastatic PCa in vivo. Furthermore, p62 deficiency triggers in adipocytes a general shutdown of energy-utilizing pathways through mTORC1 inhibition, which supports nutrient availability for cancer cells. This reveals a central role of adipocyte's p62 in the symbiotic adipose tissue-tumor collaboration that enables cancer metabolic fitness.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Obesity/complications , Osteopontin/metabolism , Prostatic Neoplasms/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Adipose Tissue/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Cell Line, Tumor , Energy Metabolism , Fatty Acids/metabolism , Humans , Male , Mice , Neoplasm Transplantation , Obesity/genetics , Obesity/metabolism , Prognosis , Prostatic Neoplasms/genetics
20.
Cell Metab ; 26(6): 817-829.e6, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-28988820

ABSTRACT

Tumors undergo nutrient stress and need to reprogram their metabolism to survive. The stroma may play a critical role in this process by providing nutrients to support the epithelial compartment of the tumor. Here we show that p62 deficiency in stromal fibroblasts promotes resistance to glutamine deprivation by the direct control of ATF4 stability through its p62-mediated polyubiquitination. ATF4 upregulation by p62 deficiency in the stroma activates glucose carbon flux through a pyruvate carboxylase-asparagine synthase cascade that results in asparagine generation as a source of nitrogen for stroma and tumor epithelial proliferation. Thus, p62 directly targets nuclear transcription factors to control metabolic reprogramming in the microenvironment and repress tumorigenesis, and identifies ATF4 as a synthetic vulnerability in p62-deficient tumor stroma.


Subject(s)
Activating Transcription Factor 4/metabolism , Cancer-Associated Fibroblasts/metabolism , Glutamine/deficiency , Prostatic Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Stress, Physiological , Tumor Microenvironment , Activating Transcription Factor 4/genetics , Animals , Asparagine/metabolism , Carcinogenesis , Cell Line, Tumor , Glucose/metabolism , HEK293 Cells , Humans , Male , Mice , RNA-Binding Proteins/genetics , Stromal Cells/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...