Network


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

Hotspot


Dive into the research topics where Colin Carlock is active.

Publication


Featured researches published by Colin Carlock.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Aurora B kinase phosphorylates and instigates degradation of p53

Chris Gully; Guermarie Velazquez-Torres; Ji Hyun Shin; Enrique Fuentes-Mattei; Edward Wang; Colin Carlock; Jian Chen; Daniel Rothenberg; Henry P. Adams; Hyun Ho Choi; Sergei Guma; Liem Phan; Ping Chieh Chou; Chun Hui Su; Fanmao Zhang; Jiun Sheng Chen; Tsung Ying Yang; Sai Ching J. Yeung; Mong Hong Lee

Aurora B is a mitotic checkpoint kinase that plays a pivotal role in the cell cycle, ensuring correct chromosome segregation and normal progression through mitosis. Aurora B is overexpressed in many types of human cancers, which has made it an attractive target for cancer therapies. Tumor suppressor p53 is a genome guardian and important negative regulator of the cell cycle. Whether Aurora B and p53 are coordinately regulated during the cell cycle is not known. We report that Aurora B directly interacts with p53 at different subcellular localizations and during different phases of the cell cycle (for instance, at the nucleus in interphase and the centromeres in prometaphase of mitosis). We show that Aurora B phosphorylates p53 at S183, T211, and S215 to accelerate the degradation of p53 through the polyubiquitination–proteasome pathway, thus functionally suppressing the expression of p53 target genes involved in cell cycle inhibition and apoptosis (e.g., p21 and PUMA). Pharmacologic inhibition of Aurora B in cancer cells with WT p53 increased p53 protein level and expression of p53 target genes to inhibit tumor growth. Together, these results define a mechanism of p53 inactivation during the cell cycle and imply that oncogenic hyperactivation or overexpression of Aurora B may compromise the tumor suppressor function of p53. We have elucidated the antineoplastic mechanism for Aurora B kinase inhibitors in cancer cells with WT p53.


Journal of the National Cancer Institute | 2014

Effects of Obesity on Transcriptomic Changes and Cancer Hallmarks in Estrogen Receptor–Positive Breast Cancer

Enrique Fuentes-Mattei; Guermarie Velazquez-Torres; Liem Phan; Fanmao Zhang; Ping Chieh Chou; Ji Hyun Shin; Hyun Ho Choi; Jiun Sheng Chen; Ruiying Zhao; Jian Chen; Chris Gully; Colin Carlock; Yuan Qi; Ya Zhang; Yun Wu; Francisco J. Esteva; Yongde Luo; Wallace L. McKeehan; Joe Ensor; Gabriel N. Hortobagyi; Lajos Pusztai; W. Fraser Symmans; Mong Hong Lee; Sai Ching Jim Yeung

Background Obesity increases the risk of cancer death among postmenopausal women with estrogen receptor–positive (ER+) breast cancer, but the direct evidence for the mechanisms is lacking. The purpose of this study is to demonstrate direct evidence for the mechanisms mediating this epidemiologic phenomenon. Methods We analyzed transcriptomic profiles of pretreatment biopsies from a prospective cohort of 137 ER+ breast cancer patients. We generated transgenic (MMTV-TGFα;A y /a) and orthotopic/syngeneic (A y /a) obese mouse models to investigate the effect of obesity on tumorigenesis and tumor progression and to determine biological mechanisms using whole-genome transcriptome microarrays and protein analyses. We used a coculture system to examine the impact of adipocytes/adipokines on breast cancer cell proliferation. All statistical tests were two-sided. Results Functional transcriptomic analysis of patients revealed the association of obesity with 59 biological functional changes (P < .05) linked to cancer hallmarks. Gene enrichment analysis revealed enrichment of AKT-target genes (P = .04) and epithelial–mesenchymal transition genes (P = .03) in patients. Our obese mouse models demonstrated activation of the AKT/mTOR pathway in obesity-accelerated mammary tumor growth (3.7- to 7.0-fold; P < .001; n = 6–7 mice per group). Metformin or everolimus can suppress obesity-induced secretion of adipokines and breast tumor formation and growth (0.5-fold, P = .04; 0.3-fold, P < .001, respectively; n = 6–8 mice per group). The coculture model revealed that adipocyte-secreted adipokines (eg, TIMP-1) regulate adipocyte-induced breast cancer cell proliferation and invasion. Metformin suppress adipocyte-induced cell proliferation and adipocyte-secreted adipokines in vitro. Conclusions Adipokine secretion and AKT/mTOR activation play important roles in obesity-accelerated breast cancer aggressiveness in addition to hyperinsulinemia, estrogen signaling, and inflammation. Metformin and everolimus have potential for therapeutic interventions of ER+ breast cancer patients with obesity.


Nature Communications | 2015

The cell cycle regulator 14-3-3σ opposes and reverses cancer metabolic reprogramming

Liem Phan; Ping Chieh Chou; Guermarie Velazquez-Torres; Ismael Samudio; Kenneth Parreno; Yaling Huang; Chieh Tseng; Thuy Vu; Chris Gully; Chun Hui Su; Edward Wang; Jian Chen; Hyun Ho Choi; Enrique Fuentes-Mattei; Ji-Hyun Shin; Christine Y. Shiang; Brian C. Grabiner; Marzenna Blonska; Stephen Skerl; Yiping Shao; Dianna Cody; Jorge Delacerda; Charles Kingsley; Douglas Webb; Colin Carlock; Zhongguo Zhou; Yun Chih Hsieh; Jae-Hyuk Lee; Andrew M. Elliott; Marc S. Ramirez

Summary Extensive reprogramming of cellular energy metabolism is a hallmark of cancer. Despite its importance, the molecular mechanism controlling this tumour metabolic shift remains not fully understood. Here we show that 14-3-3σ regulates cancer metabolic reprogramming and protects cells from tumourigenic transformation. 14-3-3σ opposes tumour-promoting metabolic programs by enhancing c-Myc poly-ubiquitination and subsequent degradation. 14-3-3σ demonstrates the suppressive impact on cancer glycolysis, glutaminolysis, mitochondrial biogenesis and other major metabolic processes of tumours. Importantly, 14-3-3σ expression levels predict overall and recurrence-free survival rates, tumour glucose uptake and metabolic gene expression in breast cancer patients. Thus, these results highlight that 14-3-3σ is an important regulator of tumour metabolism, and loss of 14-3-3σ expression is critical for cancer metabolic reprogramming. We anticipate that pharmacologically elevating the function of 14-3-3σ in tumours could be a promising direction for targeted anti-cancer metabolism therapy development in future.


Journal of Immunology | 2015

IL-33 is required for disposal of unnecessary cells during ovarian atresia through regulation of autophagy and macrophage migration

Jean Wu; Colin Carlock; Cindy Zhou; Susumu Nakae; John Hicks; Henry P. Adams; Ya Huan Lou

Physiological processes such as ovarian follicle atresia generate large amounts of unnecessary cells or tissue detritus, which needs to be disposed of rapidly. IL-33 is a member of the IL-1 cytokine gene family. Constitutive expression of IL-33 in a wide range of tissues has hinted at its role beyond immune defense. We have previously reported a close correlation between IL-33 expression patterns and ovarian atresia. In this study, we demonstrated that IL-33 is required for disposal of degenerative tissue during ovarian atresia using Il33−/− mice. Deletion of the Il33 gene impaired normal disposal of atretic follicles, resulting in massive accumulations of tissue wastes abundant with aging-related catabolic wastes such as lipofuscin. Accumulation of tissue wastes in Il33−/− mice, in turn, accelerated ovarian aging and functional decline. Thus, their reproductive life span was shortened to two thirds of that for Il33+/− littermates. IL-33 orchestrated disposal mechanism through regulation of autophagy in degenerating tissues and macrophage migration into the tissues. Our study provides direct evidence supporting an expanded role of IL-33 in tissue integrity and aging through regulating disposal of unnecessary tissues or cells.


Journal of Immunology | 2014

Unique Temporal and Spatial Expression Patterns of IL-33 in Ovaries during Ovulation and Estrous Cycle Are Associated with Ovarian Tissue Homeostasis

Colin Carlock; Jean Wu; Cindy Zhou; Kiana Tatum; Henry P. Adams; Filemon Tan; Ya Huan Lou

Ovaries are among the most active organs. Frequently occurring events such as ovulation and ovarian atresia are accompanied with tissue destruction and repairing. Critical roles of immune cells or molecules in those events have been well recognized. IL-33 is a new member of the IL-1 cytokine gene family. Recent studies suggest its roles beyond immune responses. We systemically examined its expression in ovaries for its potential roles in ovarian functions. During ovulation, a high level of IL-33 was transiently expressed, making it the most significantly upregulated immune gene. During estrous cycle, IL-33 expression levels fluctuated along with numbers of ovarian macrophages and atresia wave. Cells with nuclear form of IL-33 (nIL-33+ cells) were mostly endothelial cells of veins, either in the inner layer of theca of ovulating follicles during ovulation, or surrounding follicles during estrous cycle. Changes in number of nIL-33+ cells showed a tendency similar to that in IL-33 mRNA level during estrous cycle. However, the cell number sharply declined before a rapid increase of macrophages and a surge of atresia. The decline in nIL-33+ cell number was coincident with detection of higher level of the cytokine form of IL-33 by Western blot, suggesting a release of cytokine form of IL-33 before the surge of macrophage migration and atresia. However, IL-33 Ab, either by passive transfer or immunization, showed a limited effect on ovulation or atresia. It raises a possibility of IL-33’s role in tissue homeostasis after ovarian events, instead of a direct involvement in ovarian functions.


Reproduction | 2013

Ovarian Phagocyte Subsets and Their Distinct Tissue Distribution Patterns

Colin Carlock; Jean Wu; Cindy Zhou; April Ross; Henry P. Adams; Ya Huan Lou

Ovarian macrophages, which play critical roles in various ovarian events, are probably derived from multiple lineages. Thus, a systemic classification of their subsets is a necessary first step for determination of their functions. Utilizing antibodies to five phagocyte markers, i.e. IA/IE (major histocompatibility complex class II), F4/80, CD11b (Mac-1), CD11c, and CD68, this study investigated subsets of ovarian phagocytes in mice. Three-color immunofluorescence and flow cytometry, together with morphological observation on isolated ovarian cells, demonstrated complicated phenotypes of ovarian phagocytes. Four macrophage and one dendritic cell subset, in addition to many minor phagocyte subsets, were identified. A dendritic cell-like population with a unique phenotype of CD11c(high)IA/IE⁻F4/80⁻ was also frequently observed. A preliminary age-dependent study showed dramatic increases in IA/IE⁺ macrophages and IA/IE⁺ dendritic cells after puberty. Furthermore, immunofluorescences on ovarian sections showed that each subset displayed a distinct tissue distribution pattern. The pattern for each subset may hint to their role in an ovarian function. In addition, partial isolation of ovarian macrophage subset using CD11b antibodies was attempted. Establishment of this isolation method may have provided us a tool for more precise investigation of each subsets functions at the cellular and molecular levels.


Kidney International | 2014

Peripheral blood CD8αα+CD11c+MHC-II+CD3- cells attenuate autoimmune glomerulonephritis in rats

Jean Wu; Cindy Zhou; Julie Robertson; Colin Carlock; Ya Huan Lou

In an anti-GBM glomerulonephritis (GN) model, GN-resistant Lewis rats naturally recover from early glomerular inflammation. Here we investigated recovery mechanisms for development of a potential immunotherapy for autoimmune GN. Our previous studies suggested that glomeruli-infiltrating leukocytes with a phenotype of CD8αα+CD11c+MHC−II+CD3− (GIL CD8αα+ cells) were responsible for recovery through induction of T cell apoptosis. Now, we identified peripheral blood CD8αα+CD11c+MHC−II+CD3− cells (PBMC CD8αα+CD3− cells), which shared 9 markers with GIL CD8αα+ cells. Upon incubation, PBMC CD8αα+CD3− cells displayed a morphology resembling that of dendritic cells. Similar to GIL CD8αα+ cells, PBMC CD8αα+CD3− cells were capable of inducing T cell apoptosis in vitro. Hence, PBMC CD8αα+CD3− cells were likely the precursor of GIL CD8αα+ cells. We next tested their potential in vivo function. PBMC CD8αα+CD3− cells were able to infiltrate inflamed but not normal glomeruli. Isolated PBMC CD8αα+CD3− cells of Lewis rats were transferred into GN-prone Wistar Kyoto rats at early inflammatory stage (day 17–25). When examined at day 45, both histopathology and BUN/serum creatinine level showed significantly attenuated GN in 80% of cell recipient Wistar Kyoto rats. Separate experiments verified infiltration of transferred Lewis PBMC CD8αα+CD3− into the glomeruli, accompanied with apoptotic CD4+ T cells in the glomeruli of the recipient Wistar Kyoto rats. Thus, PBMC CD8αα+CD3− cells of Lewis rats were able to terminate ongoing autoimmune inflammation in the glomeruli.


PLOS ONE | 2018

Inter-molecular epitope spreading does not lead to extension of autoimmunity beyond target tissue in autoimmune glomerulonephritis

April Ross; Jean Wu; Colin Carlock; William Glass; Ya Huan Lou

Inter-molecular epitope spreading during autoimmune pathogenesis leads to generation of new pathogenic epitopes on other autoantigens beyond the original one. It raises an important question as whether autoimmunity extends beyond the target tissues if new epitopes are on the molecules shared with other tissues. This study is aimed addressing this question in a rat anti-glomerular basement membrane (GBM) glomerulonephritis model induced by a T cell epitope of glomerulus-specific collagen4α3. We have demonstrated inter-molecular B cell epitope spreading. Four novel epitopes were first identified by screening a phage display random peptide library against autoantibodies isolated from the GBM of immunized rats. All four epitopes were derived from GBM proteins with three from laminins and one from collagen4α4. Three out of four synthetic peptides were nephritogenic. Importantly, two peptides from lamininα1 and lamininβ1, respectively, induced severe inflammation in glomeruli but not in the interstitial tissues, despite the presence of more abundant laminins in the tubular basement membranes. Our study suggests that surrounding tissues may display a lower or altered susceptibility to autoimmune inflammation. Thus, preventing extension of autoimmune inflammation beyond the original target tissue.


Translational Psychiatry | 2017

Interleukin33 deficiency causes tau abnormality and neurodegeneration with Alzheimer-like symptoms in aged mice

Colin Carlock; Jean Wu; J Shim; Ines Moreno-Gonzalez; M R Pitcher; John Hicks; A Suzuki; J Iwata; J Quevado; Ya Huan Lou

Late-onset Alzheimer’s disease (AD) remains a medical mystery. Recent studies have linked it to impaired repair of aged neurons. Potential involvement of interleukin33 (IL33) in AD has been reported. Here we show that IL33, which was expressed by up to 75% astrocytes in the aged brains, was critical for repair of aged neurons. Mice lacking Il33 gene (Il33−/−) developed AD-like disease after 60–80 weeks, which was characterized by tau abnormality and a heavy loss of neurons/neurites in the cerebral cortex and hippocampus accompanied with cognition/memory impairment. We detected an abrupt aging surge in the cortical and hippocampal neurons at middle age (40 weeks). To counter the aging surge, wild-type mice rapidly upregulated repair of DNA double-strand breaks (DSBs) and autophagic clearance of cellular wastes in these neurons. Il33−/− mice failed to do so, but instead went on to develop rapid accumulation of abnormal tau, massive DSBs and abnormal autophagic vacuoles in these neurons. Thus, uncontrolled neuronal aging surge at middle age due to lack of IL33 resulted in neurodegeneration and late-onset AD-like symptome in Il33−/− mice. Our study also suggests that the aging surge is a time to search for biomarkers for early diagnosis of AD before massive neuron loss.


Journal of Immunology | 2016

CD8αα+MHC Class II+ Cell with the Capacity To Terminate Autoimmune Inflammation Is a Novel Antigen-Presenting NK-like Cell in Rats

Jean Wu; Colin Carlock; April Ross; Junbo Shim; Ya Huan Lou

Discovery of immune tolerance mechanisms, which inhibit pre-existing autoimmune inflammation, may provide us with new strategies for treating autoimmune diseases. We have identified a CD8αα+MHC class II+ cell with professional APC capacity during our investigation on spontaneous recovery from autoimmune glomerulonephritis in a rat model. This cell actively invades inflamed target tissue and further terminates an ongoing autoimmune inflammation by selective killing of effector autoreactive T cells. In this study, we show that this cell used a cytotoxic machinery of Ly49s+ NK cells in killing of target T cells. Thus, this CD8αα+MHC class II+ cell was a dually functional Ag-presenting NK-like (AP-NK) cell. Following its coupling with target T cells through Ag presentation, killing stimulatory receptor Ly49s6 and coreceptor CD8αα on this cell used rat nonclassic MHC class I C/E16 on the target T cells as a ligand to initiate killing. Thus, activated effector T cells with elevated expression of rat nonclassic MHC class I C/E16 were highly susceptible to the killing by the CD8αα+ AP-NK cell. Granule cytolytic perforin/granzyme C from this cell subsequently mediated cytotoxicity. Thus, inhibition of granzyme C effectively attenuated the killing. As it can recognize and eliminate effector autoreactive T cells in the inflamed target tissue, the CD8αα+ AP-NK cell not only represents a new type of immune cell involved in immune tolerance, but it also is a potential candidate for developing a cell-based therapy for pre-existing autoimmune diseases.

Collaboration


Dive into the Colin Carlock's collaboration.

Top Co-Authors

Avatar

Jean Wu

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Ya Huan Lou

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Chris Gully

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cindy Zhou

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Enrique Fuentes-Mattei

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Guermarie Velazquez-Torres

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Henry P. Adams

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jian Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Liem Phan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

April Ross

University of Texas Health Science Center at Houston

View shared research outputs
Researchain Logo
Decentralizing Knowledge