Network


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

Hotspot


Dive into the research topics where Betty L. Cabrera is active.

Publication


Featured researches published by Betty L. Cabrera.


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.


The FASEB Journal | 2011

Expression of GIV/Girdin, a metastasis-related protein, predicts patient survival in colon cancer

Mikel Garcia-Marcos; Barbara Jung; Jason Ear; Betty L. Cabrera; John M. Carethers; Pradipta Ghosh

Metastasis accounts for the majority of cancer‐related deaths. Accurate prediction of meta‐static potential of tumors has been elusive, and the search for clinically useful markers continues. We previously reported that GIV/Girdin triggers tumor cell migration by virtue of a C‐terminal guanine‐nucle‐otide exchange factor motif that activates Gai. Here we identify GIV as a metastasis‐related protein whose full‐length transcript (GIV‐fl) is expressed exclusively in highly invasive colon, breast, and pancreatic carcinoma cells and not in their poorly invasive counterparts. A prospective, exploratory biomarker study conducted on a cohort of 56 patients with stage II colorectal cancer revealed a significant correlation between GIV‐fl expression in tumor epithelium and shortened metastasis‐free survival. Survival rate for patients with GIV‐fl‐positive tumors is significantly reduced compared with the patients with GIV‐fl‐negative tumors [P<0.0001;hazard ratio=0.076;CI=0.052–0.30 (95%)]. At the 5‐yr mark, survival is 100% in the GIV‐fl‐negative group and 62 ± 9% (mean±se; P=6×10‐5) in the GIV‐fl‐positive group. Furthermore, GIV‐fl expression predicts a risk of mortality independent of the microsatellite stability status, a well‐established prognosticator of colo‐rectal cancers. We conclude that GIV‐fl is a novel metastasis‐related protein and an independent adverse prognos‐ticator that may serve as a useful adjunct to traditional staging strategies in colorectal carcinoma.—Garcia‐Marcos, M., Jung, B. H., Ear, J., Cabrera, B., Carethers, J. M., Ghosh, P. Expression of GIV/Girdin, a metastasis‐related protein, predicts patient survival in colon cancer. FASEB J. 25, 590–599 (2011). www.fasebj.org


PLOS ONE | 2014

Influence of race on microsatellite instability and CD8+ T cell infiltration in colon cancer

John M. Carethers; Bhavya Murali; Bing Yang; Ryan T. Doctolero; Akihiro Tajima; Ranor C. B. Basa; E. Julieta Smith; Monte Lee; Ryan Janke; Tina Ngo; Ruth Tejada; Ming Ji; Matthew Kinseth; Betty L. Cabrera; Katsumi Miyai; Temitope O. Keku; Christopher F. Martin; Joseph A. Galanko; Robert S. Sandler; Kathleen L. McGuire

African American patients with colorectal cancer show higher mortality than their Caucasian counterparts. Biology might play a partial role, and prior studies suggest a higher prevalence for microsatellite instability (MSI) among cancers from African Americans, albeit patients with MSI cancers have improved survival over patients with non-MSI cancers, counter to the outcome observed for African American patients. CD8+ T cell infiltration of colon cancer is postively correlated with MSI tumors, and is also related to improved outcome. Here, we utilized a 503-person, population-based colon cancer cohort comprising 45% African Americans to determine, under blinded conditions from all epidemiological data, the prevalence of MSI and associated CD8+ T cell infiltration within the cancers. Among Caucasian cancers, 14% were MSI, whereas African American cancers demonstrated 7% MSI (P = 0.009). Clinically, MSI cancers between races were similar; among microsatellite stable cancers, African American patients were younger, female, and with proximal cancers. CD8+ T cells were higher in MSI cancers (88.0 vs 30.4/hpf, P<0.0001), but was not different between races. Utilizing this population-based cohort, African American cancers show half the MSI prevalence of Caucasians without change in CD8+ T cell infiltration which may contribute towards their higher mortality from colon cancer.


Alimentary Pharmacology & Therapeutics | 2011

Enteric‐coated cysteamine for the treatment of paediatric non‐alcoholic fatty liver disease

Ranjan Dohil; Susanne Schmeltzer; Betty L. Cabrera; T. Wang; Janis Durelle; K. B. Duke; Jeffrey B. Schwimmer; Joel E. Lavine

Aliment Pharmacol Ther 2011; 33: 1036–1044


PLOS ONE | 2011

Both hMutSα and hMutSß DNA Mismatch Repair Complexes Participate in 5-Fluorouracil Cytotoxicity

Akihiro Tajima; Moriya Iwaizumi; Stephanie Tseng-Rogenski; Betty L. Cabrera; John M. Carethers

Background Patients with advanced microsatellite unstable colorectal cancers do not show a survival benefit from 5-fluorouracil (5-FU)-based chemotherapy. We and others have shown that the DNA mismatch repair (MMR) complex hMutSα binds 5-FU incorporated into DNA. Although hMutSß is known to interact with interstrand crosslinks (ICLs) induced by drugs such as cisplatin and psoralen, it has not been demonstrated to interact with 5-FU incorporated into DNA. Our aim was to examine if hMutSß plays a role in 5-FU recognition. Methods We compared the normalized growth of 5-FU treated cells containing either or both mismatch repair complexes using MTT and clonogenic assays. We utilized oligonucleotides containing 5-FU and purified baculovirus-synthesized hMutSα and hMutSß in electromobility shift assays (EMSA) and further analyzed binding using surface plasmon resonance. Results MTT and clonogenic assays after 5-FU treatment demonstrated the most cytotoxicity in cells with both hMutSα and hMutSß, intermediate cytotoxicity in cells with hMutSα alone, and the least cytotoxicity in cells with hMutSß alone, hMutSß binds 5-FU-modified DNA, but its relative binding is less than the binding of 5-FU-modified DNA by hMutSα. Conclusion Cytotoxicity induced by 5-FU is dependent on intact DNA MMR, with relative cell death correlating directly with hMutSα and/or hMutSß 5-FU binding ability (hMutSα>hMutSß). The MMR complexes provide a hierarchical chemosensitivity for 5-FU cell death, and may have implications for treatment of patients with certain MMR-deficient tumors.


The Journal of Pediatrics | 2012

The Effect of Cysteamine Bitartrate on Adiponectin Multimerization in Non-Alcoholic Fatty Liver Disease and Healthy Subjects

Ranjan Dohil; Lauren Meyer; Susanne Schmeltzer; Betty L. Cabrera; Joel E. Lavine; Susan A. Phillips

OBJECTIVE To determine the effects of cysteamine on adiponectin multimerization in sera of patients with nonalcoholic fatty liver disease (NAFLD). STUDY DESIGN Sera from 10 children with biopsy-proven NAFLD treated with cysteamine were assayed for adiponectin multimers at baseline, after 24 weeks of treatment, and again 16 weeks after discontinuing treatment. Pretreatment sera from subjects with NAFLD and from adult controls without NAFLD controls (n = 8) were incubated in cysteamine and multimers were measured 1 hour later. A cysteamine/adiponectin multimer dose-response curve was created. RESULTS Following 24 weeks of cysteamine therapy, the mean percentage increase for high, medium (MMW), and low (LMW) molecular weight multimers and total adiponectin from baseline was 53% (P = .02), 19% (P = .02), 29.4% (P = .03), and 49.3% (P = .05), respectively. Levels returned to baseline at 16 weeks after stopping therapy, unlike hepatic transaminase levels which remained low. Sera from 0 week, incubated in cysteamine for 1 hour, showed a significant mean percent increase in LMW adiponectin levels and a mean percent reduction in MMW levels compared with baseline in adults with and without NAFLD. CONCLUSIONS Cysteamine impacts adiponectin multimerization. Long-term cysteamine therapy increases levels of all multimers, whereas, in vitro short-term exposure causes a rapid increase in LMW and reduction in MMW multimers in NAFLD and healthy controls. Cysteamine may be a potential therapeutic agent for conditions associated with insulin-resistance, oxidative stress, and depressed adiponectin levels.


The Journal of Pediatrics | 2010

Long-Term Treatment of Cystinosis in Children with Twice-Daily Cysteamine

Ranjan Dohil; Jon A. Gangoiti; Betty L. Cabrera; Meredith Fidler; Jerry A. Schneider; Bruce Barshop

OBJECTIVE Cystinosis causes renal and other organ failure. Treatment with 6-hourly cysteamine bitartrate (Cystagon, Mylan, Morgantown, West Virginia) reduces intracellular cystine and the rate of organ deterioration. A recent study showed that an enteric-release cysteamine required less frequent daily dosing. This report describes the long-term use of enteric-coated (EC) cysteamine bitartrate (Cystagon) in children with cystinosis. STUDY DESIGN After a pharmacokinetic and pharmacodynamic study of EC-cysteamine in children with cystinosis, 5 patients remained on twice-daily treatment. White blood cell cystine levels were measured 12 hours after ingestion every 4 to 8 weeks. These levels were then compared with the patients previous 6-h post-dose levels taken while on regular cysteamine bitartrate before entering the study. Blood chemistry was also measured. RESULTS Five children with cystinosis (mean age, 9 years; range, 8 to 17 years) who previously took cysteamine bitartrate (mean dose, 47 mg/kg body wt), received EC-cysteamine for 10 to 27 months (mean dose, 25 mg/kg body wt) and had mean white blood cell cystine levels of 0.77 and 0.71 nmol half-cystine/mg protein, respectively. During the study period, patients maintained adequate growth and there was no significant deterioration in renal or thyroid function. Two children were required to restart acid suppression after 6 months on EC-cysteamine therapy. CONCLUSIONS Long-term, twice-daily EC-cysteamine, given at approximately 60% of the previous daily dose of cysteamine bitartrate, was effective at maintaining white blood cell cystine levels within a satisfactory range. There was no significant deterioration in renal or thyroid function.


British Journal of Clinical Pharmacology | 2010

Pharmacokinetics of enteric-coated cysteamine bitartrate in healthy adults: a pilot study

Jon A. Gangoiti; Meredith Fidler; Betty L. Cabrera; Jerry A. Schneider; Bruce Barshop; Ranjan Dohil

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT Cysteamine bitartrate is taken lifelong, every 6 h and for the treatment of cystinosis. Recent studies using cysteamine for for other diseases such as neurodegenerative disorders adopt the same dosing regimen for cysteamine. Regular cysteamine bitartrate (Cystagon) may cause upper gastrointestinal symptoms in some patients. WHAT THIS STUDY ADDS This is the only study that provides pharmacokinetic data for cysteamine delivered in an enteric-release preparation in normal subjects. EC-cysteamine is very well tolerated and does not cause increased gastrin concentrations, even at relatively high doses. EC-cysteamine at the higher dose results in better drug uptake as measured by Cmax and AUC and is more likely to be effective. AIMS Cysteamine bitartrate (Cystagon) is the approved treatment for cystinosis. Poor compliance and patient outcome may occur because the drug needs to be taken every 6 h and in some patients causes gastrointestinal symptoms due to hypergastrinaemia. A formulation of cysteamine requiring twice daily ingestion would improve the quality of life for these patients. This study compares the pharmacokinetics and gastrin production following cysteamine bitartrate non-enteric-coated and cysteamine bitartrate enteric-coated in normal healthy subjects. METHODS Enteric-coated cysteamine was prepared. Following single doses of cysteamine bitartrate non-enteric-coated 450 mg and cysteamine bitartrate enteric-coated 450 mg and 900 mg, serial plasma cysteamine and gastrin concentrations were measured. Two subjects also received cysteamine bitartrate non-enteric-coated 900 mg. Gastrointestinal (GI) symptoms were recorded. RESULTS Six healthy adults (mean age 20.7 years, range 18-24 years; mean weight 59.3 kg) received drug. All post-dose gastrin concentrations were within the normal range (<100 pg ml(-1)). The tmax following cysteamine bitartrate non-enteric-coated (mean and SD is 75+/-19 min) was shorter than cysteamine bitartrate enteric-coated (220+/-74 min) (P=0.001), but only the Cmax and AUC estimates following 900 mg cysteamine bitartrate enteric-coated were significantly greater than any of the other preparations or doses (P<0.05). One patient had GI symptoms following both 900 mg cysteamine bitartrate non-enteric-coated and cysteamine bitartrate enteric-coated. CONCLUSION Although patient numbers were low, single high doses of cysteamine bitartrate enteric-coated were better tolerated than similar doses of cysteamine bitartrate non-enteric-coated in the healthy subjects and all had normal gastrin concentrations. The delayed tmax following cysteamine bitartrate enteric-coated suggested that the cysteamine was released enterically.


Oncogene | 2010

Flanking sequence specificity determines coding microsatellite heteroduplex and mutation rates with defective DNA mismatch repair (MMR)

Heekyung Chung; Claudia G. Lopez; Dennis J. Young; Jenny F. Lai; Joy Holmstrom; Deena Ream-Robinson; Betty L. Cabrera; John M. Carethers

The activin type II receptor (ACVR2) contains two identical microsatellites in exons 3 and 10, but only the exon 10 microsatellite is frameshifted in mismatch repair (MMR)-defective colonic tumors. The reason for this selectivity is not known. We hypothesized that ACVR2 frameshifts were influenced by DNA sequences surrounding the microsatellite. We constructed plasmids in which exons 3 or 10 of ACVR2 were cloned +1 bp out of frame of enhanced green fluorescent protein (EGFP), allowing –1 bp frameshift to express EGFP. Plasmids were stably transfected into MMR-deficient cells, and subsequent non-fluorescent cells were sorted, cultured and harvested for mutation analysis. We swapped DNA sequences flanking the exon 3 and 10 microsatellites to test our hypothesis. Native ACVR2 exon 3 and 10 microsatellites underwent heteroduplex formation (A7/T8) in hMLH1−/− cells, but only exon 10 microsatellites fully mutated (A7/T7) in both hMLH1−/− and hMSH6−/− backgrounds, showing selectivity for exon 10 frameshifts and inability of exon 3 heteroduplexes to fully mutate. Substituting nucleotides flanking the exon 3 microsatellite for nucleotides flanking the exon 10 microsatellite significantly reduced heteroduplex and full mutation in hMLH1−/− cells. When the exon 3 microsatellite was flanked by nucleotides normally surrounding the exon 10 microsatellite, fully mutant exon 3 frameshifts appeared. Mutation selectivity for ACVR2 lies partly with flanking nucleotides surrounding each microsatellite.


Fundamental & Clinical Pharmacology | 2014

Pharmacokinetics of cysteamine bitartrate following intraduodenal delivery.

Ranjan Dohil; Betty L. Cabrera; Jon A. Gangoiti; Bruce Barshop; Patrice Rioux

Cysteamine is approved for the treatment of cystinosis and is being evaluated for Huntingtons disease and non‐alcoholic fatty liver disease. Little is known about the bioavailability and biodistribution of the drug. The aim was to determine plasma, cerebrospinal fluid (CSF), and tissue (liver, kidney, muscle) cysteamine levels following intraduodenal delivery of the drug in rats pretreated and naïve to cysteamine and to estimate the hepatic first‐pass effect on cysteamine. Healthy male rats (n = 66) underwent intraduodenal and portal (PV) or jugular (JVC) venous catheterization. Half were pretreated with cysteamine, and half were naïve. Following intraduodenal cysteamine (20 mg/kg), serial blood samples were collected from the PV or the JVC. Animals were sacrificed at specific time points, and CSF and tissue were collected. Cysteamine levels were determined in plasma, CSF, and tissue. The Cmax was achieved in 5–10 min from PV and 5–22.5 min from JVC. The PV‐Cmax (P = 0.08), PV‐AUC0–t (P = 0.16), JVC‐Cmax (P = 0.02) and JVC‐AUC0–t (P = 0.03) were higher in naive than in pretreated animals. Plasma cysteamine levels returned to baseline in ≤120 min. The hepatic first‐pass effect was estimated at 40%. Peak tissue and CSF cysteamine levels occurred ≤22.5 min, but returned to baseline levels ≤180 min. There was no difference in CSF and tissue cysteamine levels between naïve and pretreated groups, although cysteamine was more rapidly cleared in the pretreated group. Cysteamine is rapidly absorbed from the small intestine, undergoes significant hepatic first‐pass metabolism, crosses the blood brain barrier, and is almost undetectable in plasma, CSF, and body tissues 2 h after ingestion. Sustained‐release cysteamine may provide prolonged tissue exposure.

Collaboration


Dive into the Betty L. Cabrera's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ranjan Dohil

University of California

View shared research outputs
Top Co-Authors

Avatar

Akihiro Tajima

University of California

View shared research outputs
Top Co-Authors

Avatar

Barbara Jung

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katsumi Miyai

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stayce E. Beck

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruce Barshop

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge