Network


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

Hotspot


Dive into the research topics where Lorraine K. Morlock is active.

Publication


Featured researches published by Lorraine K. Morlock.


Journal of the American Chemical Society | 2011

Development of Proneurogenic, Neuroprotective Small Molecules

Karen S. MacMillan; Jacinth Naidoo; Jue Liang; Lisa Melito; Noelle S. Williams; Lorraine K. Morlock; Paula Huntington; Sandi Jo Estill; Jamie Longgood; Ginger L. Becker; Steven L. McKnight; Andrew A. Pieper; Jef K. De Brabander; Joseph M. Ready

Degeneration of the hippocampus is associated with Alzheimers disease and occurs very early in the progression of the disease. Current options for treating the cognitive symptoms associated with Alzheimers are inadequate, giving urgency to the search for novel therapeutic strategies. Pharmacologic agents that safely enhance hippocampal neurogenesis may provide new therapeutic approaches. We discovered the first synthetic molecule, named P7C3, which protects newborn neurons from apoptotic cell death, and thus promotes neurogenesis in mice and rats in the subgranular zone of the hippocampal dentate gyrus, the site of normal neurogenesis in adult mammals. We describe the results of a medicinal chemistry campaign to optimize the potency, toxicity profile, and stability of P7C3. Systematic variation of nearly every position of the lead compound revealed elements conducive toward increases in activity and regions subject to modification. We have discovered compounds that are orally available, nontoxic, stable in mice, rats, and cell culture, and capable of penetrating the blood-brain barrier. The most potent compounds are active at nanomolar concentrations. Finally, we have identified derivatives that may facilitate mode-of-action studies through affinity chromatography or photo-cross-linking.


Science Translational Medicine | 2012

A Validated Tumorgraft Model Reveals Activity of Dovitinib Against Renal Cell Carcinoma

Sharanya Sivanand; Samuel Peña-Llopis; Hong Zhao; Blanka Kucejova; Patrick Spence; Andrea Pavia-Jimenez; Toshinari Yamasaki; David McBride; Jessica Gillen; Nicholas C. Wolff; Lorraine K. Morlock; Yair Lotan; Ganesh V. Raj; Arthur I. Sagalowsky; Vitaly Margulis; Jeffrey A. Cadeddu; Mark T. Ross; David R. Bentley; Wareef Kabbani; Xian Jin Xie; Payal Kapur; Noelle S. Williams; James Brugarolas

Extensively validated tumorgraft model shows activity of investigational agent dovitinib against renal cell carcinoma. Grafting a Better Cancer Model When it comes to predicting drug responsiveness in cancer patients, the standard mouse models (xenografts) get low marks: Drugs that work in these mice are frequently ineffective in humans. Xenograft models are created by injecting human tumor cell lines—which often acquire new mutations in culture—into immunocompromised mice. The resulting tumors are generally different from the original tumor. Tumorgrafts, instead created by implanting fragments of human tumors directly into mice, are generating new excitement among some researchers. Sivanand et al. now describe and validate a tumorgraft model of renal cell carcinoma (RCC) that shows promise for preclinical drug studies. The scientists implanted small fragments of RCC tumor from 94 patients into mice, placing the fragments under the fibrous capsule that surrounds the kidney. Sixteen stable tumor lines—which could be serially passaged to new mice—were ultimately established. Examination by a clinical pathologist revealed that the tumorgrafts were quite similar histologically to the original tumors; gene expression patterns, DNA copy number changes, and most mutations in protein-coding regions were also preserved. In addition, mice bearing tumorgrafts from patients that developed elevated concentrations of serum calcium similarly developed tumor-induced hypercalcemia. Moreover, tumorgraft growth was inhibited by two drugs (sunitinib, approved for treating RCC, and sirolimus, the active metabolite of an approved RCC drug), but did not respond to a lung cancer drug that is inactive against RCC. Finally, dovitinib, an inhibitor of several growth factor receptors that is being studied in clinical trials, suppressed the tumorgrafts more potently than sunitinib or sirolimus, suggesting that dovitinib would be active against RCC in humans. These results apply more broadly: The tumorgrafts should be useful for testing other targeted drugs preclinically—an important need, given that most anticancer drugs that enter clinical trials fail to gain approval. Most anticancer drugs entering clinical trials fail to achieve approval from the U.S. Food and Drug Administration. Drug development is hampered by the lack of preclinical models with therapeutic predictive value. Herein, we report the development and validation of a tumorgraft model of renal cell carcinoma (RCC) and its application to the evaluation of an experimental drug. Tumor samples from 94 patients were implanted in the kidneys of mice without additives or disaggregation. Tumors from 35 of these patients formed tumorgrafts, and 16 stable lines were established. Samples from metastatic sites engrafted at higher frequency than those from primary tumors, and stable engraftment of primary tumors in mice correlated with decreased patient survival. Tumorgrafts retained the histology, gene expression, DNA copy number alterations, and more than 90% of the protein-coding gene mutations of the corresponding tumors. As determined by the induction of hypercalcemia in tumorgraft-bearing mice, tumorgrafts retained the ability to induce paraneoplastic syndromes. In studies simulating drug exposures in patients, RCC tumorgraft growth was inhibited by sunitinib and sirolimus (the active metabolite of temsirolimus in humans), but not by erlotinib, which was used as a control. Dovitinib, a drug in clinical development, showed greater activity than sunitinib and sirolimus. The routine incorporation of models recapitulating the molecular genetics and drug sensitivities of human tumors into preclinical programs has the potential to improve oncology drug development.


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

Neuroprotective efficacy of aminopropyl carbazoles in a mouse model of Parkinson disease

Héctor De Jesús-Cortés; Pin Xu; Jordan Drawbridge; Sandi Jo Estill; Paula Huntington; Stephanie Tran; Jeremiah K. Britt; Rachel Tesla; Lorraine K. Morlock; Jacinth Naidoo; Lisa Melito; Gelin Wang; Noelle S. Williams; Joseph M. Ready; Steven L. McKnight; Andrew A. Pieper

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the dentate gyrus. Here, we provide evidence that P7C3 also protects mature neurons in brain regions outside of the hippocampus. P7C3 blocks 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated cell death of dopaminergic neurons in the substantia nigra of adult mice, a model of Parkinson disease (PD). Dose–response studies show that the P7C3 analog P7C3A20 blocks cell death with even greater potency and efficacy, which parallels the relative potency and efficacy of these agents in blocking apoptosis of newborn neural precursor cells of the dentate gyrus. P7C3 and P7C3A20 display similar relative effects in blocking 1-methyl-4-phenylpyridinium (MPP+)-mediated death of dopaminergic neurons in Caenorhabditis elegans, as well as in preserving C. elegans mobility following MPP+ exposure. Dimebon, an antihistaminergic drug that is weakly proneurogenic and neuroprotective in the dentate gyrus, confers no protection in either the mouse or the worm models of PD. We further demonstrate that the hippocampal proneurogenic efficacy of eight additional analogs of P7C3 correlates with their protective efficacy in MPTP-mediated neurotoxicity. In vivo screening of P7C3 analogs for proneurogenic efficacy in the hippocampus may thus provide a reliable means of predicting neuroprotective efficacy. We propose that the chemical scaffold represented by P7C3 and P7C3A20 provides a basis for optimizing and advancing pharmacologic agents for the treatment of patients with PD.


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

Neuroprotective efficacy of aminopropyl carbazoles in a mouse model of amyotrophic lateral sclerosis

Rachel Tesla; Hamilton Parker Wolf; Pin Xu; Jordan Drawbridge; Sandi Jo Estill; Paula Huntington; Latisha McDaniel; Whitney Knobbe; Aaron Burket; Stephanie Tran; Ruth Starwalt; Lorraine K. Morlock; Jacinth Naidoo; Noelle S. Williams; Joseph M. Ready; Steven L. McKnight; Andrew A. Pieper

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the hippocampal dentate gyrus. We have further found that chemicals having efficacy in this in vivo screening assay also protect dopaminergic neurons of the substantia nigra following exposure to the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a mouse model of Parkinson disease. Here, we provide evidence that an active analog of P7C3, known as P7C3A20, protects ventral horn spinal cord motor neurons from cell death in the G93A-SOD1 mutant mouse model of amyotrophic lateral sclerosis (ALS). P7C3A20 is efficacious in this model when administered at disease onset, and protection from cell death correlates with preservation of motor function in assays of walking gait and in the accelerating rotarod test. The prototypical member of this series, P7C3, delays disease progression in G93A-SOD1 mice when administration is initiated substantially earlier than the expected time of symptom onset. Dimebon, an antihistaminergic drug with significantly weaker proneurogenic and neuroprotective efficacy than P7C3, confers no protection in this ALS model. We propose that the chemical scaffold represented by P7C3 and P7C3A20 may provide a basis for the discovery and optimization of pharmacologic agents for the treatment of ALS.


Molecular Psychiatry | 2015

The P7C3 class of neuroprotective compounds exerts antidepressant efficacy in mice by increasing hippocampal neurogenesis

Angela K. Walker; Phillip D. Rivera; Qian Wang; Jen-Chieh Chuang; Stephanie Tran; Sherri Osborne-Lawrence; Sandi Jo Estill; Ruth Starwalt; Paula Huntington; Lorraine K. Morlock; Jacinth Naidoo; Noelle S. Williams; Joseph M. Ready; Amelia J. Eisch; Andrew A. Pieper; Jeffrey M. Zigman

Augmenting hippocampal neurogenesis represents a potential new strategy for treating depression. Here we test this possibility by comparing hippocampal neurogenesis in depression-prone ghrelin receptor (Ghsr)-null mice to that in wild-type littermates and by determining the antidepressant efficacy of the P7C3 class of neuroprotective compounds. Exposure of Ghsr-null mice to chronic social defeat stress (CSDS) elicits more severe depressive-like behavior than in CSDS-exposed wild-type littermates, and exposure of Ghsr-null mice to 60% caloric restriction fails to elicit antidepressant-like behavior. CSDS resulted in more severely reduced cell proliferation and survival in the ventral dentate gyrus (DG) subgranular zone of Ghsr-null mice than in that of wild-type littermates. Also, caloric restriction increased apoptosis of DG subgranular zone cells in Ghsr-null mice, although it had the opposite effect in wild-type littermates. Systemic treatment with P7C3 during CSDS increased survival of proliferating DG cells, which ultimately developed into mature (NeuN+) neurons. Notably, P7C3 exerted a potent antidepressant-like effect in Ghsr-null mice exposed to either CSDS or caloric restriction, while the more highly active analog P7C3-A20 also exerted an antidepressant-like effect in wild-type littermates. Focal ablation of hippocampal stem cells with radiation eliminated this antidepressant effect, further attributing the P7C3 class antidepressant effect to its neuroprotective properties and resultant augmentation of hippocampal neurogenesis. Finally, P7C3-A20 demonstrated greater proneurogenic efficacy than a wide spectrum of currently marketed antidepressant drugs. Taken together, our data confirm the role of aberrant hippocampal neurogenesis in the etiology of depression and suggest that the neuroprotective P7C3-compounds represent a novel strategy for treating patients with this disease.


Journal of Biological Chemistry | 2014

Structure-guided Development of Specific Pyruvate Dehydrogenase Kinase Inhibitors Targeting the ATP-binding Pocket

Shih Chia Tso; Xiangbing Qi; Wen Jun Gui; Cheng Yang Wu; Jacinta L. Chuang; Ingrid Wernstedt-Asterholm; Lorraine K. Morlock; Kyle R. Owens; Philipp E. Scherer; Noelle S. Williams; Uttam K. Tambar; R. Max Wynn; David T. Chuang

Background: Up-regulated pyruvate dehydrogenase kinase isoforms (PDKs) are associated with impaired glucose homeostasis in diabetes. Results: Novel PDK inhibitors were developed using structure-based design, which improves glucose tolerance with reduced hepatic steatosis in diet-induced obese mice. Conclusion: Obesity phenotypes are effectively treated by chemical intervention with PDK inhibitors. Significance: PDKs are potential drug targets for obesity and type 2 diabetes. Pyruvate dehydrogenase kinase isoforms (PDKs 1–4) negatively regulate activity of the mitochondrial pyruvate dehydrogenase complex by reversible phosphorylation. PDK isoforms are up-regulated in obesity, diabetes, heart failure, and cancer and are potential therapeutic targets for these important human diseases. Here, we employed a structure-guided design to convert a known Hsp90 inhibitor to a series of highly specific PDK inhibitors, based on structural conservation in the ATP-binding pocket. The key step involved the substitution of a carbonyl group in the parent compound with a sulfonyl in the PDK inhibitors. The final compound of this series, 2-[(2,4-dihydroxyphenyl)sulfonyl]isoindoline-4,6-diol, designated PS10, inhibits all four PDK isoforms with IC50 = 0.8 μm for PDK2. The administration of PS10 (70 mg/kg) to diet-induced obese mice significantly augments pyruvate dehydrogenase complex activity with reduced phosphorylation in different tissues. Prolonged PS10 treatments result in improved glucose tolerance and notably lessened hepatic steatosis in the mouse model. The results support the pharmacological approach of targeting PDK to control both glucose and fat levels in obesity and type 2 diabetes.


Neuroscience | 2015

PHARMACOLOGIC RESCUE OF MOTOR AND SENSORY FUNCTION BY THE NEUROPROTECTIVE COMPOUND P7C3 FOLLOWING NEONATAL NERVE INJURY

S.W.P. Kemp; M. Szynkaruk; K.N. Stanoulis; M.D. Wood; E.H. Liu; M.P. Willand; Lorraine K. Morlock; Jacinth Naidoo; Noelle S. Williams; Joseph M. Ready; Thomas J. Mangano; S. Beggs; M.W. Salter; Tessa Gordon; Andrew A. Pieper; G.H. Borschel

Nerve injuries cause pain, paralysis and numbness that can lead to major disability, and newborns often sustain nerve injuries during delivery that result in lifelong impairment. Without a pharmacologic agent to enhance functional recovery from these injuries, clinicians rely solely on surgery and rehabilitation to treat patients. Unfortunately, patient outcomes remain poor despite application of the most advanced microsurgical and rehabilitative techniques. We hypothesized that the detrimental effects of traumatic neonatal nerve injury could be mitigated with pharmacologic neuroprotection, and tested whether the novel neuroprotective agent P7C3 would block peripheral neuron cell death and enhance functional recovery in a rat neonatal nerve injury model. Administration of P7C3 after sciatic nerve crush injury doubled motor and sensory neuron survival, and also promoted axon regeneration in a dose-dependent manner. Treatment with P7C3 also enhanced behavioral and muscle functional recovery, and reversed pathological mobilization of spinal microglia after injury. Our findings suggest that the P7C3 family of neuroprotective compounds may provide a basis for the development of a new neuroprotective drug to enhance recovery following peripheral nerve injury.


Journal of Medicinal Chemistry | 2014

Discovery of a Neuroprotective Chemical, (S)-N-(3-(3,6-Dibromo-9H-carbazol-9-yl)-2-fluoropropyl)-6-methoxypyridin-2-amine [(−)-P7C3-S243], with Improved Druglike Properties

Jacinth Naidoo; Héctor De Jesús-Cortés; Paula Huntington; Sandi Jo Estill; Lorraine K. Morlock; Ruth Starwalt; Thomas J. Mangano; Noelle S. Williams; Andrew A. Pieper; Joseph M. Ready

(−)-P7C3-S243 is a neuroprotective aminopropyl carbazole with improved druglike properties compared with previously reported compounds in the P7C3 class. It protects developing neurons in a mouse model of hippocampal neurogenesis and protects mature neurons within the substantia nigra in a mouse model of Parkinson’s disease. A short, enantioselective synthesis provides the neuroprotective agent in optically pure form. It is nontoxic, orally bioavailable, metabolically stable, and able to cross the blood–brain barrier. As such, it represents a valuable lead compound for the development of drugs to treat neurodegenerative diseases and traumatic brain injury.


Investigative Ophthalmology & Visual Science | 2014

Early detection of subclinical visual damage after blast-mediated TBI enables prevention of chronic visual deficit by treatment with P7C3-S243

Laura M. Dutca; Steven F. Stasheff; Adam Hedberg-Buenz; Danielle S. Rudd; Nikhil Batra; Frederick R. Blodi; Matthew S. Yorek; Terry Yin; Malini Shankar; Judith A. Herlein; Jacinth Naidoo; Lorraine K. Morlock; Noelle S. Williams; Randy H. Kardon; Michael G. Anderson; Andrew A. Pieper; Matthew M. Harper

PURPOSE Traumatic brain injury (TBI) frequently leads to chronic visual dysfunction. The purpose of this study was to investigate the effect of TBI on retinal ganglion cells (RGCs), and to test whether treatment with the novel neuroprotective compound P7C3-S243 could prevent in vivo functional deficits in the visual system. METHODS Blast-mediated TBI was modeled using an enclosed over-pressure blast chamber. The RGC physiology was evaluated using a multielectrode array and pattern electroretinogram (PERG). Histological analysis of RGC dendritic field and cell number were evaluated at the end of the study. Visual outcome measures also were evaluated based on treatment of mice with P7C3-S243 or vehicle control. RESULTS We show that deficits in neutral position PERG after blast-mediated TBI occur in a temporally bimodal fashion, with temporary recovery 4 weeks after injury followed by chronically persistent dysfunction 12 weeks later. This later time point is associated with development of dendritic abnormalities and irreversible death of RGCs. We also demonstrate that ongoing pathologic processes during the temporary recovery latent period (including abnormalities of RGC physiology) lead to future dysfunction of the visual system. We report that modification of PERG to provocative postural tilt testing elicits changes in PERG measurements that correlate with a key in vitro measures of damage: the spontaneous and light-evoked activity of RGCs. Treatment with P7C3-S243 immediately after injury and throughout the temporary recovery latent period protects mice from developing chronic visual system dysfunction. CONCLUSIONS Provocative PERG testing serves as a noninvasive test in the living organism to identify early damage to the visual system, which may reflect corresponding damage in the brain that is not otherwise detectable by noninvasive means. This provides the basis for developing an earlier diagnostic test to identify patients at risk for developing chronic CNS and visual system damage after TBI at an earlier stage when treatments may be more effective in preventing these sequelae. In addition, treatment with the neuroprotective agent P7C3-S243 after TBI protects from visual system dysfunction after TBI.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Structure-based design and mechanisms of allosteric inhibitors for mitochondrial branched-chain α-ketoacid dehydrogenase kinase

Shih Chia Tso; Xiangbing Qi; Wen Jun Gui; Jacinta L. Chuang; Lorraine K. Morlock; Amy L. Wallace; Kamran Ahmed; Sunil Laxman; Philippe M. Campeau; Brendan Lee; Susan M. Hutson; Benjamin P. Tu; Noelle S. Williams; Uttam K. Tambar; Richard M Wynn; David T. Chuang

The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are elevated in maple syrup urine disease, heart failure, obesity, and type 2 diabetes. BCAA homeostasis is controlled by the mitochondrial branched-chain α-ketoacid dehydrogenase complex (BCKDC), which is negatively regulated by the specific BCKD kinase (BDK). Here, we used structure-based design to develop a BDK inhibitor, (S)-α-chloro-phenylpropionic acid [(S)-CPP]. Crystal structures of the BDK-(S)-CPP complex show that (S)-CPP binds to a unique allosteric site in the N-terminal domain, triggering helix movements in BDK. These conformational changes are communicated to the lipoyl-binding pocket, which nullifies BDK activity by blocking its binding to the BCKDC core. Administration of (S)-CPP to mice leads to the full activation and dephosphorylation of BCKDC with significant reduction in plasma BCAA concentrations. The results buttress the concept of targeting mitochondrial BDK as a pharmacological approach to mitigate BCAA accumulation in metabolic diseases and heart failure.

Collaboration


Dive into the Lorraine K. Morlock's collaboration.

Top Co-Authors

Avatar

Noelle S. Williams

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrew A. Pieper

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jacinth Naidoo

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joseph M. Ready

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Paula Huntington

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sandi Jo Estill

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

David T. Chuang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jacinta L. Chuang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shih Chia Tso

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wen Jun Gui

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge