Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Patrick A. Carroll is active.

Publication


Featured researches published by Patrick A. Carroll.


Cancer Cell | 2015

Deregulated Myc Requires MondoA/Mlx for Metabolic Reprogramming and Tumorigenesis

Patrick A. Carroll; Daniel Diolaiti; Lisa McFerrin; Haiwei Gu; Danijel Djukovic; Jianhai Du; Pei Feng Cheng; Sarah Anderson; Michelle Ulrich; James B. Hurley; Daniel Raftery; Donald E. Ayer; Robert N. Eisenman

Deregulated Myc transcriptionally reprograms cell metabolism to promote neoplasia. Here we show that oncogenic Myc requires the Myc superfamily member MondoA, a nutrient-sensing transcription factor, for tumorigenesis. Knockdown of MondoA, or its dimerization partner Mlx, blocks Myc-induced reprogramming of multiple metabolic pathways, resulting in apoptosis. Identification and knockdown of genes coregulated by Myc and MondoA have allowed us to define metabolic functions required by deregulated Myc and demonstrate a critical role for lipid biosynthesis in survival of Myc-driven cancer. Furthermore, overexpression of a subset of Myc and MondoA coregulated genes correlates with poor outcome of patients with diverse cancers. Coregulation of cancer metabolism by Myc and MondoA provides the potential for therapeutics aimed at inhibiting MondoA and its target genes.


Biochimica et Biophysica Acta | 2015

Functional Interactions Among Members of the MAX and MLX Transcriptional Network During Oncogenesis

Daniel Diolaiti; Lisa McFerrin; Patrick A. Carroll; Robert N. Eisenman

The transcription factor MYC and its related family members MYCN and MYCL have been implicated in the etiology of a wide spectrum of human cancers. Compared to other oncoproteins, such as RAS or SRC, MYC is unique because its protein coding region is rarely mutated. Instead, MYCs oncogenic properties are unleashed by regulatory mutations leading to unconstrained high levels of expression. Under both normal and pathological conditions MYC regulates multiple aspects of cellular physiology including proliferation, differentiation, apoptosis, growth and metabolism by controlling the expression of thousands of genes. How a single transcription factor exerts such broad effects remains a fascinating puzzle. Notably, MYC is part of a network of bHLHLZ proteins centered on the MYC heterodimeric partner MAX and its counterpart, the MAX-like protein MLX. This network includes MXD1-4, MNT, MGA, MONDOA and MONDOB proteins. With some exceptions, MXD proteins have been functionally linked to cell cycle arrest and differentiation, while MONDO proteins control cellular metabolism. Although the temporal expression patterns of many of these proteins can differ markedly they are frequently expressed simultaneously in the same cellular context, and potentially bind to the same, or similar DNA consensus sequence. Here we review the activities and interactions among these proteins and propose that the broad spectrum of phenotypes elicited by MYC deregulation is intimately connected to the functions and regulation of the other network members. Furthermore, we provide a meta-analysis of TCGA data suggesting that the coordinate regulation of the network is important in MYC driven tumorigenesis. This article is part of a Special Issue entitled: Myc proteins in cell biology and pathology.


Cell Stem Cell | 2015

Quantification of Retinogenesis in 3D Cultures Reveals Epigenetic Memory and Higher Efficiency in iPSCs Derived from Rod Photoreceptors

Daniel Hiler; Xiang Chen; Jennifer L. Hazen; Sergey Kupriyanov; Patrick A. Carroll; Chunxu Qu; Beisi Xu; Dianna A. Johnson; Lyra Griffiths; Sharon Frase; Alberto R. Rodriguez; Greg Martin; Jiakun Zhang; Jongrye Jeon; Yiping Fan; David Finkelstein; Robert N. Eisenman; Kristin K. Baldwin; Michael A. Dyer

Cell-based therapies to treat retinal degeneration are now being tested in clinical trials. However, it is not known whether the source of stem cells is important for the production of differentiated cells suitable for transplantation. To test this, we generated induced pluripotent stem cells (iPSCs) from murine rod photoreceptors (r-iPSCs) and scored their ability to make retinae by using a standardized quantitative protocol called STEM-RET. We discovered that r-iPSCs more efficiently produced differentiated retinae than did embryonic stem cells (ESCs) or fibroblast-derived iPSCs (f-iPSCs). Retinae derived from f-iPSCs had fewer amacrine cells and other inner nuclear layer cells. Integrated epigenetic analysis showed that DNA methylation contributes to the defects in f-iPSC retinogenesis and that rod-specific CTCF insulator protein-binding sites may promote r-iPSC retinogenesis. Together, our data suggest that the source of stem cells is important for producing retinal neurons in three-dimensional (3D) organ cultures.


PLOS Pathogens | 2015

Latent KSHV Infected Endothelial Cells Are Glutamine Addicted and Require Glutaminolysis for Survival

Erica L. Sanchez; Patrick A. Carroll; Angel B. Thalhofer; Michael Lagunoff

Kaposi’s Sarcoma-associated Herpesvirus (KSHV) is the etiologic agent of Kaposi’s Sarcoma (KS). KSHV establishes a predominantly latent infection in the main KS tumor cell type, the spindle cell, which is of endothelial cell origin. KSHV requires the induction of multiple metabolic pathways, including glycolysis and fatty acid synthesis, for the survival of latently infected endothelial cells. Here we demonstrate that latent KSHV infection leads to increased levels of intracellular glutamine and enhanced glutamine uptake. Depletion of glutamine from the culture media leads to a significant increase in apoptotic cell death in latently infected endothelial cells, but not in their mock-infected counterparts. In cancer cells, glutamine is often required for glutaminolysis to provide intermediates for the tri-carboxylic acid (TCA) cycle and support for the production of biosynthetic and bioenergetic precursors. In the absence of glutamine, the TCA cycle intermediates alpha-ketoglutarate (αKG) and pyruvate prevent the death of latently infected cells. Targeted drug inhibition of glutaminolysis also induces increased cell death in latently infected cells. KSHV infection of endothelial cells induces protein expression of the glutamine transporter, SLC1A5. Chemical inhibition of SLC1A5, or knockdown by siRNA, leads to similar cell death rates as glutamine deprivation and, similarly, can be rescued by αKG. KSHV also induces expression of the heterodimeric transcription factors c-Myc-Max and related heterodimer MondoA-Mlx. Knockdown of MondoA inhibits expression of both Mlx and SLC1A5 and induces a significant increase in cell death of only cells latently infected with KSHV, again, fully rescued by the supplementation of αKG. Therefore, during latent infection of endothelial cells, KSHV activates and requires the Myc/MondoA-network to upregulate the glutamine transporter, SLC1A5, leading to increased glutamine uptake for glutaminolysis. These findings expand our understanding of the required metabolic pathways that are activated during latent KSHV infection of endothelial cells, and demonstrate a novel role for the extended Myc-regulatory network, specifically MondoA, during latent KSHV infection.


Angewandte Chemie | 2016

Quantitative Method to Investigate the Balance between Metabolism and Proteome Biomass: Starting from Glycine.

Haiwei Gu; Patrick A. Carroll; Jianhai Du; Jiangjiang Zhu; Fausto Carnevale Neto; Robert N. Eisenman; Daniel Raftery

The balance between metabolism and biomass is very important in biological systems; however, to date there has been no quantitative method to characterize the balance. In this methodological study, we propose to use the distribution of amino acids in different domains to investigate this balance. It is well known that endogenous or exogenous amino acids in a biological system are either metabolized or incorporated into free amino acids (FAAs) or proteome amino acids (PAAs). Using glycine (Gly) as an example, we demonstrate a novel method to accurately determine the amounts of amino acids in various domains using serum, urine, and cell samples. As expected, serum and urine had very different distributions of FAA- and PAA-Gly. Using Tet21N human neuroblastoma cells, we also found that Myc(oncogene)-induced metabolic reprogramming included a higher rate of metabolizing Gly, which provides additional evidence that the metabolism of proliferating cells is adapted to facilitate producing new cells. It is therefore anticipated that our method will be very valuable for further studies of the metabolism and biomass balance that will lead to a better understanding of human cancers.


Analyst | 2015

Metabolomics method to comprehensively analyze amino acids in different domains.

Haiwei Gu; Jianhai Du; Fausto Carnevale Neto; Patrick A. Carroll; Sally J. Turner; E. Gabriela Chiorean; Robert N. Eisenman; Daniel Raftery

Amino acids play essential roles in both metabolism and the proteome. Many studies have profiled free amino acids (FAAs) or proteins; however, few have connected the measurement of FAA with individual amino acids in the proteome. In this study, we developed a metabolomics method to comprehensively analyze amino acids in different domains, using two examples of different sample types and disease models. We first examined the responses of FAAs and insoluble-proteome amino acids (IPAAs) to the Myc oncogene in Tet21N human neuroblastoma cells. The metabolic and proteomic amino acid profiles were quite different, even under the same Myc condition, and their combination provided a better understanding of the biological status. In addition, amino acids were measured in 3 domains (FAAs, free and soluble-proteome amino acids (FSPAAs), and IPAAs) to study changes in serum amino acid profiles related to colon cancer. A penalized logistic regression model based on the amino acids from the three domains had better sensitivity and specificity than that from each individual domain. To the best of our knowledge, this is the first study to perform a combined analysis of amino acids in different domains, and indicates the useful biological information available from a metabolomics analysis of the protein pellet. This study lays the foundation for further quantitative tracking of the distribution of amino acids in different domains, with opportunities for better diagnosis and mechanistic studies of various diseases.


Genes & Development | 2015

The glucose-sensing transcription factor MLX promotes myogenesis via myokine signaling

Liam Hunt; Beisi Xu; David Finkelstein; Yiping Fan; Patrick A. Carroll; Pei-Feng Cheng; Robert N. Eisenman; Fabio Demontis

Metabolic stress and changes in nutrient levels modulate many aspects of skeletal muscle function during aging and disease. Growth factors and cytokines secreted by skeletal muscle, known as myokines, are important signaling factors, but it is largely unknown whether they modulate muscle growth and differentiation in response to nutrients. Here, we found that changes in glucose levels increase the activity of the glucose-responsive transcription factor MLX (Max-like protein X), which promotes and is necessary for myoblast fusion. MLX promotes myogenesis not via an adjustment of glucose metabolism but rather by inducing the expression of several myokines, including insulin-like growth factor 2 (IGF2), whereas RNAi and dominant-negative MLX reduce IGF2 expression and block myogenesis. This phenotype is rescued by conditioned medium from control muscle cells and by recombinant IGF2, which activates the myogenic kinase Akt. Importantly, MLX-null mice display decreased IGF2 induction and diminished muscle regeneration in response to injury, indicating that the myogenic function of MLX is manifested in vivo. Thus, glucose is a signaling molecule that regulates myogenesis and muscle regeneration via MLX/IGF2/Akt signaling.


Frontiers of Medicine in China | 2018

The MYC transcription factor network: balancing metabolism, proliferation and oncogenesis

Patrick A. Carroll; Brian Freie; Haritha Mathsyaraja; Robert N. Eisenman

Transcription factor networks have evolved in order to control, coordinate, and separate, the functions of distinct network modules spatially and temporally. In this review we focus on the MYC network (also known as the MAX-MLX Network), a highly conserved super-family of related basic-helix-loop-helix-zipper (bHLHZ) proteins that functions to integrate extracellular and intracellular signals and modulate global gene expression. Importantly the MYC network has been shown to be deeply involved in a broad spectrum of human and other animal cancers. Here we summarize molecular and biological properties of the network modules with emphasis on functional interactions among network members. We suggest that these network interactions serve to modulate growth and metabolism at the transcriptional level in order to balance nutrient demand with supply, to maintain growth homeostasis, and to influence cell fate. Moreover, oncogenic activation of MYC and/or loss of a MYC antagonist, results in an imbalance in the activity of the network as a whole, leading to tumor initiation, progression and maintenance.


Functional Genomics | 2018

PO-346 The MYC antagonist MNT beyond MAX interaction

Mc Lafita-Navarro; J Liano-Pons; Ignacio Varela; R Blanco; J Aresti; Patrick A. Carroll; Pj Hurlin; Robert N. Eisenman; Delgado; Javier León

Introduction MNT has been described as an antagonist and modulator of MYC, one of the most prevalent oncoproteins in human cancer. Both MYC and MNT are bHLH-LZ transcription factors that heterodimerize with MAX, bind to E-boxes within regulatory regions of target genes, and generally activate (MYC) or repress (MNT) their transcription. Material and methods The cell lines used, URMT and URMax34, derive from MAX-deficient PC12 (rat pheochromocytoma), and carry a pHeBo-MT (empty vector) and a pHeBo-MT-MAX vector (MAX-inducible with Zn+2), respectively. Knockdown of MNT and MLX were achieved with short hairpin RNA constructs (shMNT and shMLX). Proliferation was assessed by cell counting and clonogenic assays; subG0-G1 population was determined by flow cytometry. RNA-seq was performed from two experiments of MNT silencing in URMT and URMax34 cells and confirmed by RT-qPCR. Changes in protein levels were analysed by western blot. Co-immunoprecipitation and proximity ligation assays were used to study protein-protein interactions. Results and discussions Knocking-down of MNT in UR61 cells resulted in an important decrease in cell proliferation, together with a decrease in both survivin and cyclin A, which are markers of pro-survival and cell proliferation, respectively. DNA content was measured by flow cytometry, revealing an increase in sub-G0 population in shMNT cells. Thus, MNT is required for optimal proliferation of these cells. This is the first evidence of a MAX-independent function of MNT. Then, we extracted RNA from two experiments of MNT silenced and carried out RNA-seq. This resulted in 158 genes whose expression was altered. Cell cycle, DNA replication and DNA repair genes were downregulated upon MNT silencing. However, there were other up-regulated genes like the cell cycle inhibitor CDKN1C (p57). As we confirmed gene regulation by MNT without MAX, we wondered whether it could be working as an heterodimer with MLX or as an homodimer. Co-immunoprecipitation and proximity-ligation assays showed MNT’s ability to form homodimers and heterodimers with MLX. Finally, we carried out MLX knockdown and determined the genes regulated by MNT-MLX or MNT-MNT complexes. Conclusion In summary, we report novel MAX-independent functions of MNT. In our MAX-deficient model, MNT can be found in homodimers (MNT-MNT) or heterodimers (MNT-MLX) and it supports proliferation and regulates cell cycle and DNA repair genes. This new data about MNT can open new insights into cell biology and tumour development promoted by MYC.


bioRxiv | 2017

The MYC antagonist MNT autoregulates its expression and supports proliferation in MAX deficient cells

M. Carmen Lafita-Navarro; Judit Liaño-Pons; Andrea Quintanilla; Ignacio Varela; Fabiana Ourique; Gabriel Bretones; Rosa Blanco; Julia Aresti; Patrick A. Carroll; Peter J. Hurlin; Robert N. Eisenman; M. Dolores Delgado; Javier León

MNT is a transcription factor of the MXD family. MNT-MAX dimers down-regulate genes by binding to E-boxes sequences, which can also be bound by MYC-MAX to activate transcription. MNT has been described as a modulator of MYC activity but little is known about MNT regulation and whether MNT has MAX-independent functions. Using a MAX deficient cell line and siRNA-mediated silencing of MAX, we show that in the absence of MAX the total MNT levels are elevated and that MNT localizes both in the cytoplasm and the nucleus. In contrast, MNT is predominantly nuclear when MAX is expressed. MNT is required for optimal cell proliferation even in the absence of MAX, being the first report of a MAX-independent function of MNT. Interestingly, MNT forms homodimers and autoregulates its expression by repressing its own promoter. The tight MNT regulation and its activity in absence of MAX suggest its importance on cell homeostasis.

Collaboration


Dive into the Patrick A. Carroll's collaboration.

Top Co-Authors

Avatar

Robert N. Eisenman

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Daniel Raftery

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Haiwei Gu

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Daniel Diolaiti

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jianhai Du

West Virginia University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donald E. Ayer

Huntsman Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

E. Gabriela Chiorean

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lisa McFerrin

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge