Gregory C. Sartor
University of Miami
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Gregory C. Sartor.
Frontiers in Genetics | 2012
Gregory C. Sartor; Georges St. Laurent; Claes Wahlestedt
Prolonged drug use causes long-lasting neuroadaptations in reward-related brain areas that contribute to addiction. Despite significant amount of research dedicated to understanding the underlying mechanisms of addiction, the molecular underpinnings remain unclear. At the same time, much of the pervasive transcription that encompasses the human genome occurs in the nervous system and contributes to its heterogeneity and complexity. Recent evidence suggests that non-coding RNAs (ncRNAs) play an important and dynamic role in transcriptional regulation, epigenetic signaling, stress response, and plasticity in the nervous system. Dysregulation of ncRNAs are thought to contribute to many, and perhaps all, neurological disorders, including addiction. Here, we review recent insights in the functional relevance of ncRNAs, including both microRNAs (miRNAs), and long non-coding RNAs, and then illustrate specific examples of ncRNA regulation in the context of drug addiction. We conclude that ncRNAs are importantly involved in the persistent neuroadaptations associated with addiction-related behaviors, and that therapies that target specific ncRNAs may represent new avenues for the treatment of drug addiction.
The Journal of Neuroscience | 2015
Gregory C. Sartor; Samuel K. Powell; Claes Wahlestedt
Epigenetic processes that regulate histone acetylation play an essential role in behavioral and molecular responses to cocaine. To date, however, only a small fraction of the mechanisms involved in the addiction-associated acetylome have been investigated. Members of the bromodomain and extraterminal (BET) family of epigenetic “reader” proteins (BRD2, BRD3, BRD4, and BRDT) bind acetylated histones and serve as a scaffold for the recruitment of macromolecular complexes to modify chromatin accessibility and transcriptional activity. The role of BET proteins in cocaine-induced plasticity, however, remains elusive. Here, we used behavioral, pharmacological, and molecular techniques to examine the involvement of BET bromodomains in cocaine reward. Of the BET proteins, BRD4, but not BRD2 or BRD3, was significantly elevated in the nucleus accumbens (NAc) of mice and rats following repeated cocaine injections and self-administration. Systemic and intra-accumbal inhibition of BRD4 with the BET inhibitor, JQ1, attenuated the rewarding effects of cocaine in a conditioned place preference procedure but did not affect conditioned place aversion, nor did JQ1 alone induce conditioned aversion or preference. Investigating the underlying mechanisms, we found that repeated cocaine injections enhanced the binding of BRD4, but not BRD3, to the promoter region of Bdnf in the NAc, whereas systemic injection of JQ1 attenuated cocaine-induced expression of Bdnf in the NAc. JQ1 and siRNA-mediated knockdown of BRD4 in vitro also reduced expression of Bdnf. These findings indicate that disrupting the interaction between BET proteins and their acetylated lysine substrates may provide a new therapeutic avenue for the treatment of drug addiction. SIGNIFICANCE STATEMENT Proteins involved in the “readout” of lysine acetylation marks, referred to as BET bromodomain proteins (including BRD2, BRD3, BRD4, and BRDT), have been shown to be key regulators of chromatin dynamics and disease, and BET inhibitors are currently being studied in several clinical trials. However, their role in addiction-related phenomena remains unknown. In the current studies, we revealed that BRD4 is elevated in the nucleus accumbens and recruited to promoter regions of addiction-related genes following repeated cocaine administration, and that inhibition of BRD4 attenuates transcriptional and behavioral responses to cocaine. Together, these studies reveal that BET inhibitors may have therapeutic utility in the treatment of cocaine addiction.
Current Alzheimer Research | 2016
Marco Magistri; Dmitry Velmeshev; Madina Makhmutova; Prutha Patel; Gregory C. Sartor; Claude Henry Volmar; Claes Wahlestedt; Mohammad Ali Faghihi
Abstract: Background Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by well-defined neuropathological brain changes including amyloid plaques, neurofibrillary tangles and the presence of chronic neuroinflammation. Objective: The brain penetrant BET bromodomain inhibitor JQ1 has been shown to regulate inflammation responses in vitro and in vivo, but its therapeutic potential in AD is currently unknown. Method: Three-month-old 3xTg mice were injected once a day with JQ1 (50 mg/kg) or vehicle for 15 weeks. At the end of the treatment learning and memory was assessed using the modified Barnes maze and the Y maze behavioral tests. Tissue from the brain and other organs was collected for molecular evaluation of neuroinflammation tau pathology and amyloid β. Results: JQ1 treatment reduced splenomegaly and neuroinflammation in the brain of treated mice where we observed a reduction in the expression of the pro-inflammatory modulators Il-1b, Il-6, Tnfa, Ccl2, Nos2 and Ptgs2. Additionally, JQ1-treated mice showed a reduction of tau phosphorylation at Ser396 in the hippocampus and frontal cortex while total levels of tau remained unaffected. On the other hand, JQ1 did not ameliorate learning and memory deficits in 7-month-old 3xTg mice. Conclusion: Taken together, our data suggest that BET bromodomain inhibitors hold the promise to be used for the treatment of neurological disorders characterized by neuroinflammation.
Brain Research | 2016
Gregory C. Sartor; Samuel K. Powell; H.J. Wiedner; Claes Wahlestedt
Growing evidence indicates that targeting nociceptin receptor (NOP) signaling may have therapeutic efficacy in treating alcohol and opioid addiction. However, little is known about the therapeutic value of selective NOP agonists for the treatment of cocaine dependence. Recently, we identified a highly selective, brain-penetrant NOP small molecule agonist (SR-8993), and using this compound, we previously showed that nociceptin receptor activation attenuated consolidation of fear-related memories. Here, we sought to determine whether SR-8993 also affects the rewarding properties of cocaine. Using a conditioned place preference (CPP) procedure, we show that SR-8993 (3 or 10 mg/kg) failed to disrupt acquisition or expression of cocaine CPP (7.5 or 15 mg/kg) in C57BL/6 mice. Additionally, SR-8993 did not affect rate of extinction or reinstatement (yohimbine- and cocaine-induced) of cocaine CPP. These studies indicate that selective activation of NOP may not be sufficient in reducing behavioral responses to cocaine.
Proceedings of the National Academy of Sciences of the United States of America | 2017
Claude Henry Volmar; Hasib Salah-Uddin; Karolina J. Janczura; Paul Halley; Guerline Lambert; Andrew Wodrich; Sivan Manoah; Nidhi H. Patel; Gregory C. Sartor; Neil Mehta; Nancy T.H. Miles; Sachi Desse; David Dorcius; Michael D. Cameron; Claes Wahlestedt
Significance Hundreds of failed clinical trials with Alzheimer’s disease (AD) patients over the last fifteen years demonstrate that the one-target–one-disease approach is not effective in AD. In silico, structure-based, multitarget drug design approaches to treat multifactorial diseases have not been successful in the context of AD either. Here, we show that M344, an inhibitor of class I and IIB histone deacetylases, affects multiple AD-related genes, including those related to both early- and late-onset AD. We also show that M344 improves memory in the 3xTg AD mouse model. This work endorses a shift to a multitargeted approach to the treatment of AD, supporting the therapeutic potential of a single small molecule with an epigenetic mechanism of action. Alzheimer’s disease (AD) comprises multifactorial ailments for which current therapeutic strategies remain insufficient to broadly address the underlying pathophysiology. Epigenetic gene regulation relies upon multifactorial processes that regulate multiple gene and protein pathways, including those involved in AD. We therefore took an epigenetic approach where a single drug would simultaneously affect the expression of a number of defined AD-related targets. We show that the small-molecule histone deacetylase inhibitor M344 reduces beta-amyloid (Aβ), reduces tau Ser396 phosphorylation, and decreases both β-secretase (BACE) and APOEε4 gene expression. M344 increases the expression of AD-relevant genes: BDNF, α-secretase (ADAM10), MINT2, FE65, REST, SIRT1, BIN1, and ABCA7, among others. M344 increases sAPPα and CTFα APP metabolite production, both cleavage products of ADAM10, concordant with increased ADAM10 gene expression. M344 also increases levels of immature APP, supporting an effect on APP trafficking, concurrent with the observed increase in MINT2 and FE65, both shown to increase immature APP in the early secretory pathway. Chronic i.p. treatment of the triple transgenic (APPsw/PS1M146V/TauP301L) mice with M344, at doses as low as 3 mg/kg, significantly prevented cognitive decline evaluated by Y-maze spontaneous alternation, novel object recognition, and Barnes maze spatial memory tests. M344 displays short brain exposure, indicating that brief pulses of daily drug treatment may be sufficient for long-term efficacy. Together, these data show that M344 normalizes several disparate pathogenic pathways related to AD. M344 therefore serves as an example of how a multitargeting compound could be used to address the polygenic nature of multifactorial diseases.
Hypertension | 2018
Wei Zhu; Huilan Wang; Jianqin Wei; Gregory C. Sartor; Michelle Meiqi Bao; Clay T. Pierce; Claes Wahlestedt; Derek M. Dykxhoorn; Chunming Dong
Cocaine abuse increases the risk of cardiovascular mortality and morbidity; however, the underlying molecular mechanisms remain elusive. By using a mouse model for cocaine abuse/use, we found that repeated cocaine injection led to increased blood pressure and aortic stiffness in mice associated with elevated levels of reactive oxygen species (ROS) in the aortas, a phenomenon similar to that observed in hypertensive humans. This ROS elevation was correlated with downregulation of Me1 (malic enzyme 1), an important redox molecule that counteracts ROS generation, and upregulation of microRNA (miR)-30c-5p that targets Me1 expression by directly binding to its 3′UTR (untranslated region). Remarkably, lentivirus-mediated overexpression of miR-30c-5p in aortic smooth muscle cells recapitulated the effect of cocaine on Me1 suppression, which in turn led to ROS elevation. Moreover, in vivo silencing of miR-30c-5p in smooth muscle cells resulted in Me1 upregulation, ROS reduction, and significantly suppressed cocaine-induced increases in blood pressure and aortic stiffness—a similar effect to that produced by treatment with the antioxidant N-acetyl cysteine. Discovery of this novel cocaine-↑miR-30c-5p–↓Me1–↑ROS pathway provides a potential new therapeutic avenue for treatment of cocaine abuse–related cardiovascular disease.
Molecular and Cellular Neuroscience | 2017
Gregory C. Sartor; Samuel K. Powell; Dmitry Velmeshev; David Y. Lin; Marco Magistri; Hannah J. Wiedner; Andrea M. Malvezzi; Nadja S. Andrade; Mohammad Ali Faghihi; Claes Wahlestedt
ABSTRACT Natural antisense transcripts (NATs) are an abundant class of long noncoding RNAs that have recently been shown to be key regulators of chromatin dynamics and gene expression in nervous system development and neurological disorders. However, it is currently unclear if NAT‐based mechanisms also play a role in drug‐induced neuroadaptations. Aberrant regulation of gene expression is one critical factor underlying the long‐lasting behavioral abnormalities that characterize substance use disorder, and it is possible that some drug‐induced transcriptional responses are mediated, in part, by perturbations in NAT activity. To test this hypothesis, we used an automated algorithm that mines the NCBI AceView transcriptomics database to identify NAT overlapping genes linked to addiction. We found that 22% of the genes examined contain NATs and that expression of Homer1 natural antisense transcript (Homer1‐AS) was altered in the nucleus accumbens (NAc) of mice 2 h and 10 days following repeated cocaine administration. In in vitro studies, depletion of Homer1‐AS lead to an increase in the corresponding sense gene expression, indicating a potential regulatory mechanisms of Homer1 expression by its corresponding antisense transcript. Future in vivo studies are needed to definitely determine a role for Homer1‐AS in cocaine‐induced behavioral and molecular adaptations. HighlightsMany genes that have been implicated in addiction overlap with natural antisense transcripts.Homer1 antisense transcript is significantly increased in the nucleus accumbens by cocaine.Knockdown of Homer1 antisense transcript increases expression of Homer1 in vitro.
Alzheimers & Dementia | 2018
Claude-Henry Volmar; Karolina J. Janczura; Guerline Lambert; Hasib Salah-Uddin; Paul Halley; Gregory C. Sartor; Claes Wahlestedt
Background:Recently, a panel of experts gathered by the National Institute of Aging and the Alzheimer’s Association published new biological guidelines for the definition of Alzheimer’s disease (AD) rather than the symptomatic approach typically used in the clinic. Such a change was judged necessary, considering that no FDA-approved AD treatments currently address the main hallmarks of the disease. With the recent clinical trial shortcomings of AD immunotherapy, gand b-secretase inhibitors, among other interventions, we and colleagues agree that a multifactorial approach is needed to address the polygenicity of AD. We used an epigenetic strategy where a single drug would simultaneously affect the expression of well-defined AD-related targets. Methods: AD-related genes and proteins were analyzed using NanoString technology, RT-qPCR methods, Western blots and ELISAs (N 1⁄4 3 to 6). Behavioral effects on the 3xTg AD mouse model were assessed using the open field, the Y-maze, the Barnes Maze and the novel object recognition tests (N1⁄410). Unpaired Student’s T-test was used whenever only two means were being compared. Oneway ANOVA or repeated measures two-way ANOVAwith appropriate post hoc analyses were used for multiple comparisons. Results:Pan and selective HDAC inhibition both reduce Ab(1-42) accumulation (p<0.05), decrease tau phosphorylation (p<0.01), and increase the expression of several high-priority AD-related protective genes. For example, compound M344 normalizes late-onset risk factor AD genes such as APOEe4 (p<0.0001) and BIN1 (p<0.0001). M344 also results in prevention of cognitive impairment in the 3xTg AD mice. M344 shows low toxicity, and rapidly clears out of brain and plasma. Similar positive results are also obtained with a more selective HDAC inhibitor compound. Conclusions:Our data suggest that brief daily brain exposure of a small epigenetic molecule can target the non-amyloidogenic pathway, increase neuroprotective genes, show low toxicity and increase memory in an AD model. This work supports a shift to a multitargeted approach to the treatment of AD.
Psychopharmacology | 2016
Abdul Maruf Asif Aziz; Gregory C. Sartor; Lovisa Holm; Markus Heilig; Claes Wahlestedt; Annika Thorsell
Biological Psychiatry | 2018
Claude-Henry Volmar; Hasib Salah-Uddin; Karolina J. Janczura; Paul Halley; Guerline Lambert; Andrew Wodrich; Sivan Manoah; Nidhi H. Patel; Gregory C. Sartor; Neil Mehta; Nancy T.H. Miles; Sachi Desse; David Dorcius; Michael D. Cameron; Claes Wahlestedt