Network


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

Hotspot


Dive into the research topics where John M. Carethers is active.

Publication


Featured researches published by John M. Carethers.


Gastroenterology | 2008

Genomic and Epigenetic Instability in Colorectal Cancer Pathogenesis

William M. Grady; John M. Carethers

Colorectal cancer arises as a consequence of the accumulation of genetic alterations (gene mutations, gene amplification, and so on) and epigenetic alterations (aberrant DNA methylation, chromatin modifications, and so on) that transform colonic epithelial cells into colon adenocarcinoma cells. The loss of genomic stability and resulting gene alterations are key molecular pathogenic steps that occur early in tumorigenesis; they permit the acquisition of a sufficient number of alterations in tumor suppressor genes and oncogenes that transform cells and promote tumor progression. Two predominant forms of genomic instability that have been identified in colon cancer are microsatellite instability and chromosome instability. Substantial progress has been made to identify causes of chromosomal instability in colorectal cells and to determine the effects of the different forms of genomic instability on the biological and clinical behavior of colon tumors. In addition to genomic instability, epigenetic instability results in the aberrant methylation of tumor suppressor genes. Determining the causes and roles of genomic and epigenomic instability in colon tumor formation has the potential to yield more effective prevention strategies and therapeutics for patients with colorectal cancer.


Gastroenterology | 1999

Mismatch repair proficiency and in vitro response to 5-fluorouracil☆☆☆

John M. Carethers; Dharam P. Chauhan; Daniel Fink; Sibylle Nebel; Robert S. Bresalier; Stephen B. Howell; C. Richard Boland

BACKGROUND & AIMS The DNA mismatch repair (MMR) system recognizes certain DNA adducts caused by alkylation damage in addition to its role in recognizing and directing repair of interstrand nucleotide mismatches and slippage mistakes at microsatellite sequences. Because defects in the MMR system can confer tolerance to acquired DNA damage and, by inference, the toxic effects of certain chemotherapeutic agents, we investigated the effect of 5-fluorouracil (5-FU) on colon cancer cell lines. METHODS We determined growth selection by cell enrichment assay and cloning efficiency after treatment with 5 micromol/L 5-FU, assayed nucleic 3H-5-FU incorporation, and analyzed the cell cycle by flow cytometry. RESULTS 5-FU treatment provided a growth advantage for MMR-deficient cell lines, indicating a relative degree of tolerance to 5-FU by the MMR-deficient cell lines. Enhanced survival was statistically significant after 5 days of growth, and a 28-fold reduction in survival was noted in the MMR-proficient cells by clonagenic assays after 10 days of growth. Differences in nucleotide uptake of 5-FU did not account for the observed growth differences, and specific cell cycle checkpoint arrest was not detected. CONCLUSIONS Intact DNA MMR seems to recognize 5-FU incorporated into DNA but may do so in a different manner than other types of alkylation damage. Defective DNA MMR might be one mechanism for tumor resistance to 5-FU.


Familial Cancer | 2008

The biochemical basis of microsatellite instability and abnormal immunohistochemistry and clinical behavior in Lynch Syndrome: from bench to bedside

C. Richard Boland; Minoru Koi; Dong K. Chang; John M. Carethers

Lynch syndrome is an inherited disease caused by a germline mutation in one of four DNA mismatch repair (MMR) genes. The clinical manifestations can be somewhat variable depending upon which gene is involved, and where the mutation occurs. Moreover, the approach to the diagnosis of Lynch syndrome is becoming more complex as more is learned about the disease, and one needs to understand how the DNA MMR proteins function, and what makes them malfunction, to have an optimal appreciation of how to interpret diagnostic studies such as microsatellite instability and immunohistochemistry of the DNA MMR proteins. Finally, an understanding of the role of the DNA MMR system in regulation of the cell cycle and the response to DNA damage helps illuminate the differences in natural history and response to chemotherapeutic agents seen in Lynch syndrome.


Journal of Clinical Investigation | 1996

Competency in mismatch repair prohibits clonal expansion of cancer cells treated with N-methyl-N'-nitro-N-nitrosoguanidine.

John M. Carethers; Mary T. Hawn; Dharam P. Chauhan; M C Luce; Giancarlo Marra; Minoru Koi; C R Boland

The phenomenon of alkylation tolerance has been observed in cells that are deficient in some component of the DNA mismatch repair (MMR) system. An alkylation-induced cell cycle arrest had been reported previously in one MMR-proficient cell line, whereas a MMR-defective clone derived from this line escapes from this arrest. We examined human cancer cell lines to determine if the cell cycle arrest were dependent upon the MMR system. Growth characteristics and cell cycle analysis after MNNG treatment were ascertained in seven MMR-deficient and proficient cell lines, with and without confirmed mutations in hMLH1 or hMSH2 by an in vitro transcription/translation assay. MMR-proficient cells underwent growth arrest in the G2 phase of the cell cycle after the first S phase, whereas MMR-deficient cells escaped an initial G2 delay and resumed a normal growth pattern. In the HCT116 line corrected for defective MMR by chromosome 3 transfer, the G2 phase arrest lasted more than five days. In another MMR-proficient colon cancer cell line, SW480, cell death occurred five days after MNNG treatment. A competent MMR system appears to be necessary for G2 arrest or cell death after alkylation damage, and this cell cycle checkpoint may allow the cell to repair damaged DNA, or prevent the replication of mutated DNA by prohibiting clonal expansion.


Molecular Biology of the Cell | 2010

A Gαi-GIV molecular complex binds epidermal growth factor receptor and determines whether cells migrate or proliferate

Pradipta Ghosh; Anthony Beas; Scott J. Bornheimer; Mikel Garcia-Marcos; Erin P. Forry; Carola Johannson; Jason Ear; Barbara Jung; Betty L. Cabrera; John M. Carethers; Marilyn G. Farquhar

Migrating cells do not proliferate and vice versa, but the mechanism involved remains unknown. Ghosh et al. reveal how this cellular decision is made by showing that a Gαi–GIV molecular complex interacts with EGF receptor and programs growth factor signaling, triggering migration when assembled and favoring mitosis when assembly is prevented.


Gastroenterology | 2000

Colorectal cancer prevention and treatment.

C. Richard Boland; Frank A. Sinicrope; Dean E. Brenner; John M. Carethers

Gastrointestinal cancer has attracted the attention of many basic scientists and clinical investigators over the past 2‐3 decades, and the progress made is currently being translated into new types of clinical management for our patients. This review will focus on colorectal cancer, highlight some of the newer findings that are available, and attempt to predict where future breakthroughs are likely to occur. Drug interventions directed at established cancer and novel preventive programs will be highlighted; patients are increasingly interested in understanding how they can prevent cancers in both high-risk and ordinary-risk situations. General Management Strategies for Gastrointestinal Cancer The gastrointestinal tract represents the interface with our diet. We should be mindful that the digestive organs account for more cancer than any other system. A key message is that manipulation of diet, gut flora, and the gastrointestinal milieu are reasonable targets to modify the risk of cancer in the gastrointestinal tract. Our approaches to the management of cancer and cancer risk might be broadly divided into the following categories. At the preventive end of the spectrum, we try to identify healthy people who are at increased risk for cancer and intervene with treatment programs that will prevent neoplastic lesions from developing. Some individuals who are completely healthy may approach us and ask how to reduce their risk of cancer. In the near future, we may be asked to develop cancer prevention programs


Gastroenterology | 2015

Genetics and Genetic Biomarkers in Sporadic Colorectal Cancer

John M. Carethers; Barbara H. Jung

Sporadic colorectal cancer (CRC) is a somatic genetic disease in which pathogenesis is influenced by the local colonic environment and the patients genetic background. Consolidating the knowledge of genetic and epigenetic events that occur with initiation, progression, and metastasis of sporadic CRC has identified some biomarkers that might be utilized to predict behavior and prognosis beyond staging, and inform treatment approaches. Modern next-generation sequencing of sporadic CRCs has confirmed prior identified genetic alterations and has classified new alterations. Each patients CRC is genetically unique, propelled by 2-8 driver gene alterations that have accumulated within the CRC since initiation. Commonly observed alterations across sporadic CRCs have allowed classification into a (1) hypermutated group that includes defective DNA mismatch repair with microsatellite instability and POLE mutations in ∼15%, containing multiple frameshifted genes and BRAF(V600E); (2) nonhypermutated group with multiple somatic copy number alterations and aneuploidy in ∼85%, containing oncogenic activation of KRAS and PIK3CA and mutation and loss of heterozygosity of tumor suppressor genes, such as APC and TP53; (3) CpG island methylator phenotype CRCs in ∼20% that overlap greatly with microsatellite instability CRCs and some nonhypermutated CRCs; and (4) elevated microsatellite alterations at selected tetranucleotide repeats in ∼60% that associates with metastatic behavior in both hypermutated and nonhypermutated groups. Components from these classifications are now used as diagnostic, prognostic, and treatment biomarkers. Additional common biomarkers may come from genome-wide association studies and microRNAs among other sources, as well as from the unique alteration profile of an individual CRC to apply a precision medicine approach to care.


Cancer Epidemiology, Biomarkers & Prevention | 2005

Diet, Lifestyle, and Genomic Instability in the North Carolina Colon Cancer Study

Jessie A. Satia; Temitope O. Keku; Joseph A. Galanko; Christopher Martin; Ryan T. Doctolero; Akihiro Tajima; Robert S. Sandler; John M. Carethers

Objective: Microsatellite instability (MSI) is one form of genomic instability that occurs in 10% to 20% of sporadic colon tumors and almost all hereditary nonpolyposis colon cancers. However, little is known about how environmental factors (e.g., diet) may influence MSI in sporadic colon cancer. Methods: We used data from a population-based case-control study in North Carolina (486 colon cancer cases and 1,048 controls) to examine associations of diet (total energy, macronutrients, micronutrients, and food groups) with MSI. In-person interviews elicited information on potential colon cancer risk factors, and a previously validated food frequency questionnaire adapted to include regional foods was used to assess diet over the year before diagnosis or interview date. MSI was classified as MSI-high (MSI-H) and MSI-low or microsatellite stable (MSI-L/MSS). Multivariate logistic regression models estimated energy-adjusted and non-energy-adjusted odds ratios (OR). Results: Ten percent of the cases (n = 49) had MSI-H tumors (29% African American). The strongest associations between diet and MSI were observed in case-control comparisons: there was a robust inverse association between MSI-H status and β-carotene [OR, 0.4; 95% confidence interval (95% CI), 0.2-0.9] and positive associations with energy-adjusted refined carbohydrates (OR, 2.2; 95% CI, 0.9-5.4) and non-energy-adjusted read meat intake (OR, 2.0; 95% CI, 0.9-4.2). Compared with controls, MSI-L/MSS tumors were statistically significantly associated with energy-adjusted vitamin C, vitamin E, calcium, dietary fiber, and dark green vegetables and positively associated with total energy intake (all Ps for trend < 0.05). In case-case comparisons, no dietary factors were significantly differently related to MSI-H compared with MSI-L/MSS tumors. Conclusion: Refined carbohydrate and red meat consumption may promote development of MSI-H tumors, whereas β-carotene may be associated with lower risk.


Gastroenterology | 1995

In vitro transcription/translation assay for the screening of hMLH1 and hMSH2 mutations in familial colon cancer

Michael C. Luce; Giancarlo Marra; Dharam P. Chauhan; Luigi Laghi; John M. Carethers; Sajeev P. Cherian; Mary T. Hawn; Cameron G. Binnie; Lauren Kam-Morgan; Matthew C. Cayouette; Minoru Koi; C. Richard Boland

BACKGROUND & AIMS Hereditary nonpolyposis colorectal cancer (HNPCC) has been linked recently to a defect in repairing mismatched nucleotides in DNA. The aim of this study was to screen for germline mutations that result in prematurely truncated proteins in two of the mismatch repair genes identified at this time, hMLH1 and hMSH2, in a consecutive series of patients belonging to familial aggregations of colorectal cancer. METHODS Nineteen individuals with colorectal cancer from 19 families were consecutively referred because of a strong positive family history of colorectal cancer. Premature truncation mutations in hMLH1 and hMSH2 were sought from lymphocyte RNA by using an in vitro transcription/translation (IVTT) assay. RESULTS Protein truncating mutations in the hMLH1 or hMSH2 genes were found in 50% of families with HNPCC (6 of 12) but were not observed in any of the remaining familial aggregations that did not fulfill the standard criteria for HNPCC. In some of the IVTT-positive samples, the mutations were characterized by genomic sequencing. CONCLUSIONS IVTT may be a practical method to accomplish primary screening of germline mutations in DNA mismatch pair genes in HNPCC; however, a broader approach is necessary to obtain a more complete picture of the mutational spectrum in HNPCC and other familial aggregations of colorectal cancer.


International Journal of Cancer | 2005

Clinicopathological features and microsatellite instability (MSI) in colorectal cancers from African Americans.

Hassan Ashktorab; Duane T. Smoot; Haleh Farzanmehr; Marie Fidelia-Lambert; Bahram Momen; Linda M. Hylind; Christine Iacosozio-Dononue; John M. Carethers; Ajay Goel; C. Richard Boland; Francis M. Giardiello

African Americans (AAs) have a 1.5 times higher risk of colorectal carcinoma (CRC) than Caucasians. Gene silencing through CpG island hypermethylation has been associated with the genesis or progression of microsatellite instability (MSI) largely due to 1 target for hypermethylation being the DNA mismatch repair gene hMLH1; there is anecdotal evidence of an increased incidence of MSI among AAs. P16 and hMLH1 can be inactivated by hypermethylation of their respective promoter regions, abrogating the ability to regulate cell proliferation and repair processes. We studied such methylation, as well as hMHS2 expression in colorectal cancers from AA patients to determine if MSI is associated with epigenetic silencing. Experiments were conducted on matched normal and colon cancer tissues from AA patients (n = 51). A total of 5 microsatellite markers (D2S123, D5S346, D17S250, BAT25 and BAT26) were used to evaluate MSI status. P16 and hMLH1 promoter methylation status was determined following bisulfite modification of DNA and using methylation specific PCR, while immunohistochemistry (IHC) was used to examine expression of hMLH1 and hMSH2. A total of 22 (43%) cancers demonstrated microsatellite instability‐high (MSI‐H), while 27 were microsatellite stable (MSS) and 2 were microsatellite instability‐low (MSH‐L). Most of the MSI‐H tumors were proximal, well differentiated and highly mucinous. Most patients in the MSI‐H group were females (68%). The p16 promoter was methylated in 19 of 47 (40%) tumors. A total of 7 of these CRCs demonstrated MSI‐H (33%). The hMLH1 promoter was methylated in 29 of 34 (85%) tumors, of which 13 CRCs demonstrated MSI‐H (87%). hMLH1 and hMSH2 staining was observed in 66% and 38% of MSI‐H tumors, respectively. Overall, the prevalence of MSI‐H colorectal tumor was 2‐3‐fold higher, while the defect in the percentage expression of mismatch repair (MMR) genes (hMLH1 and hMSH2) was similar in AA patients compared to the U.S. Caucasian population. Similar numbers of AA MSS tumors with p16 and hMLH1 methylation likely indicate hemimethylation of genes that might reflect environmental or genetic influences that might be more common in the AA population.

Collaboration


Dive into the John M. Carethers's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Richard Boland

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Barbara Jung

University of California

View shared research outputs
Top Co-Authors

Avatar

Minoru Koi

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Heekyung Chung

University of California

View shared research outputs
Top Co-Authors

Avatar

Ajay Goel

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Dong

University of California

View shared research outputs
Top Co-Authors

Avatar

Katsumi Miyai

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge