Network


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

Hotspot


Dive into the research topics where Major Gooyit is active.

Publication


Featured researches published by Major Gooyit.


PLOS ONE | 2013

Selective Inhibition of Matrix Metalloproteinase-9 Attenuates Secondary Damage Resulting from Severe Traumatic Brain Injury

Orr Hadass; Brittany N. Tomlinson; Major Gooyit; Shanyan Chen; Justin J. Purdy; Jennifer M. Walker; Chunyang Zhang; Andrew Giritharan; Whitley Purnell; Christopher R. Robinson; Dmitriy Shin; Valerie A. Schroeder; Mark A. Suckow; Agnes Simonyi; Grace Y. Sun; Shahriar Mobashery; Jiankun Cui; Mayland Chang; Zezong Gu

Traumatic brain injury (TBI) is a leading cause of death and long-term disability. Following the initial insult, severe TBI progresses to a secondary injury phase associated with biochemical and cellular changes. The secondary injury is thought to be responsible for the development of many of the neurological deficits observed after TBI and also provides a window of opportunity for therapeutic intervention. Matrix metalloproteinase-9 (MMP-9 or gelatinase B) expression is elevated in neurological diseases and its activation is an important factor in detrimental outcomes including excitotoxicity, mitochondrial dysfunction and apoptosis, and increases in inflammatory responses and astrogliosis. In this study, we used an experimental mouse model of TBI to examine the role of MMP-9 and the therapeutic potential of SB-3CT, a mechanism-based gelatinase selective inhibitor, in ameliorating the secondary injury. We observed that activation of MMP-9 occurred within one day following TBI, and remained elevated for 7 days after the initial insult. SB-3CT effectively attenuated MMP-9 activity, reduced brain lesion volumes and prevented neuronal loss and dendritic degeneration. Pharmacokinetic studies revealed that SB-3CT and its active metabolite, p-OH SB-3CT, were rapidly absorbed and distributed to the brain. Moreover, SB-3CT treatment mitigated microglial activation and astrogliosis after TBI. Importantly, SB-3CT treatment improved long-term neurobehavioral outcomes, including sensorimotor function, and hippocampus-associated spatial learning and memory. These results demonstrate that MMP-9 is a key target for therapy to attenuate secondary injury cascades and that this class of mechanism-based gelatinase inhibitor–with such desirable pharmacokinetic properties–holds considerable promise as a potential pharmacological treatment of TBI.


ACS Chemical Biology | 2014

A Chemical Biological Strategy to Facilitate Diabetic Wound Healing

Major Gooyit; Zhihong Peng; William R. Wolter; Hualiang Pi; Derong Ding; Dusan Hesek; Mijoon Lee; Bill Boggess; Matthew M. Champion; Mark A. Suckow; Shahriar Mobashery; Mayland Chang

A complication of diabetes is the inability of wounds to heal in diabetic patients. Diabetic wounds are refractory to healing due to the involvement of activated matrix metalloproteinases (MMPs), which remodel the tissue resulting in apoptosis. There are no readily available methods that identify active unregulated MMPs. With the use of a novel inhibitor-tethered resin that binds exclusively to the active forms of MMPs, coupled with proteomics, we quantified MMP-8 and MMP-9 in a mouse model of diabetic wounds. Topical treatment with a selective MMP-9 inhibitor led to acceleration of wound healing, re-epithelialization, and significantly attenuated apoptosis. In contrast, selective pharmacological inhibition of MMP-8 delayed wound healing, decreased re-epithelialization, and exhibited high apoptosis. The MMP-9 activity makes the wounds refractory to healing, whereas that of MMP-8 is beneficial. The treatment of diabetic wounds with a selective MMP-9 inhibitor holds great promise in providing heretofore-unavailable opportunities for intervention of this disease.


Journal of Medicinal Chemistry | 2011

Selective Water-Soluble Gelatinase Inhibitor Prodrugs

Major Gooyit; Mijoon Lee; Valerie A. Schroeder; Masahiro Ikejiri; Mark A. Suckow; Shahriar Mobashery; Mayland Chang

SB-3CT (1), a selective and potent thiirane-based gelatinase inhibitor, is effective in animal models of cancer metastasis and stroke; however, it is limited by poor aqueous solubility and extensive metabolism. We addressed these issues by blocking the primary site of metabolism and capitalizing on a prodrug strategy to achieve >5000-fold increased solubility. The amide prodrugs were quantitatively hydrolyzed in human blood to a potent gelatinase inhibitor, ND-322 (3). The arginyl amide prodrug (ND-478, 5d) was metabolically stable in mouse, rat, and human liver microsomes. Both 5d and 3 were nonmutagenic in the Ames II mutagenicity assay. The prodrug 5d showed moderate clearance of 0.0582 L/min/kg, remained mostly in the extracellular fluid compartment (Vd = 0.0978 L/kg), and had a terminal half-life of >4 h. The prodrug 5d had superior pharmacokinetic properties than those of 3, making the thiirane class of selective gelatinase inhibitors suitable for intravenous administration in the treatment of acute gelatinase-dependent diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Acceleration of diabetic wound healing using a novel protease–anti-protease combination therapy

Ming Gao; Trung T. Nguyen; Mark A. Suckow; William R. Wolter; Major Gooyit; Shahriar Mobashery; Mayland Chang

Significance Chronic wounds in diabetic patients are a devastating complication of diabetes that can lead to amputations or even death. Our work in db/db mice shows that matrix metalloproteinase (MMP)-9 contributes to delayed or impaired wound healing and that MMP-8 is involved in repairing the wound. A combination of a selective inhibitor of MMP-9 (a small molecule) and exogenously applied active recombinant MMP-8 (an enzyme) accelerates diabetic wound healing in mice. Nonhealing chronic wounds are major complications of diabetes resulting in >70,000 annual lower-limb amputations in the United States alone. The reasons the diabetic wound is recalcitrant to healing are not fully understood, and there are limited therapeutic agents that could accelerate or facilitate its repair. We previously identified two active forms of matrix metalloproteinases (MMPs), MMP-8 and MMP-9, in the wounds of db/db mice. We argued that the former might play a role in the body’s response to wound healing and that the latter is the pathological consequence of the disease with detrimental effects. Here we demonstrate that the use of compound ND-336, a novel highly selective inhibitor of gelatinases (MMP-2 and MMP-9) and MMP-14, accelerates diabetic wound healing by lowering inflammation and by enhancing angiogenesis and re-epithelialization of the wound, thereby reversing the pathological condition. The detrimental role of MMP-9 in the pathology of diabetic wounds was confirmed further by the study of diabetic MMP-9–knockout mice, which exhibited wounds more prone to healing. Furthermore, topical administration of active recombinant MMP-8 also accelerated diabetic wound healing as a consequence of complete re-epithelialization, diminished inflammation, and enhanced angiogenesis. The combined topical application of ND-336 (a small molecule) and the active recombinant MMP-8 (an enzyme) enhanced healing even more, in a strategy that holds considerable promise in healing of diabetic wounds.


ACS Chemical Neuroscience | 2012

Selective gelatinase inhibitor neuroprotective agents cross the blood-brain barrier.

Major Gooyit; Mark A. Suckow; Valerie A. Schroeder; William R. Wolter; Shahriar Mobashery; Mayland Chang

SB-3CT, a potent and selective inhibitor of matrix metalloproteinase-2 and -9, has shown efficacy in several animal models of neurological diseases. One of the greatest challenges in the development of therapeutics for neurological diseases is the inability of drugs to cross the blood-brain barrier. A sensitive bioanalytical method based on ultraperformance liquid chromatography with multiple-reaction monitoring detection was developed to measure levels of SB-3CT, its active metabolite, the α-methyl analogue, and its p-hydroxy metabolite in plasma and brain. The compounds are rapidly absorbed and are readily distributed to the brain. The pharmacokinetic properties of these gelatinase inhibitors and the efficacy shown by SB-3CT in animal models of stroke, subarachnoid hemorrhage, and spinal cord injury indicate that this class of compounds holds considerable promise in the treatment of diseases of the central nervous system.


Journal of Medicinal Chemistry | 2013

O-phenyl carbamate and phenyl urea thiiranes as selective matrix metalloproteinase-2 inhibitors that cross the blood-brain barrier.

Major Gooyit; Wei Song; Kiran V. Mahasenan; Katerina Lichtenwalter; Mark A. Suckow; Valerie A. Schroeder; William R. Wolter; Shahriar Mobashery; Mayland Chang

Brain metastasis occurs in 20-40% of cancer patients. Treatment is mostly palliative, and the inability of most drugs to penetrate the brain presents one of the greatest challenges in the development of therapeutics for brain metastasis. Matrix metalloproteinase-2 (MMP-2) plays important roles in invasion and vascularization of the central nervous system and represents a potential target for treatment of brain metastasis. Carbonate, O-phenyl carbamate, urea, and N-phenyl carbamate derivatives of SB-3CT, a selective and potent gelatinase inhibitor, were synthesized and evaluated. The O-phenyl carbamate and urea variants were selective and potent inhibitors of MMP-2. Carbamate 5b was metabolized to the potent gelatinase inhibitor 2, which was present at therapeutic concentrations in the brain. In contrast, phenyl urea 6b crossed the blood-brain barrier, however, higher doses would result in therapeutic brain concentrations. Carbamate 5b and urea 6b show potential for intervention of MMP-2-dependent diseases such as brain metastasis.


MedChemComm | 2015

Substituted 4-hydroxy-1,2,3-triazoles: synthesis, characterization and first drug design applications through bioisosteric modulation and scaffold hopping approaches

Agnese Chiara Pippione; Franco Dosio; Alex Ducime; Antonella Federico; Katia Martina; Stefano Sainas; Major Gooyit; Kim D. Janda; Donatella Boschi; Marco L. Lolli

Bioisosterism and scaffold hopping are two widely used approaches in medicinal chemistry for the purpose of lead optimization. The study highlights the physicochemical properties of the 4-hydroxy-1,2,3-triazole scaffold, a less investigated heterocyclic system. Synthetic strategies to obtain different N-substituted 4-hydroxy-1,2,3-triazole isomers are presented, and their role as possible isosteres of the carboxylic acid is discussed. The aim is to use this system to modulate the acidic moieties present in lead compounds and, at the same time, to regiodirect substituents in set directions, through targeted substitution on the three nitrogenatoms of the triazole ring. Through this approach, compounds having enhanced binding affinity, will be sought. Two examples of bioisosteric applications of this moiety are presented. In the first example, a classical bioisosteric approach mimicking the distal (S)-glutamic acid carboxyl group using the 4-hydroxy-1,2,3-triazole moiety is applied, to obtain two promising glutamate analogs. In the second example, a scaffold hopping approach is applied, replacing the phenolic moiety present in MDG-1-33A, a potent inhibitor of Onchocerca volvulus chitinase, with the 4-hydroxy-1,2,3-triazole scaffold. The 4-hydroxy-1,2,3-triazole system appears to be useful and versatile in drug design.


ACS Chemical Neuroscience | 2015

Water-Soluble MMP-9 Inhibitor Reduces Lesion Volume after Severe Traumatic Brain Injury

Mijoon Lee; Zhenzhou Chen; Brittany N. Tomlinson; Major Gooyit; Dusan Hesek; María Raquel Juárez; Rasheeq Nizam; Bill Boggess; Elena Lastochkin; Valerie A. Schroeder; William R. Wolter; Mark A. Suckow; Jiancun Cui; Shahriar Mobashery; Zezong Gu; Mayland Chang

SB-3CT is a potent and selective inhibitor of matrix metalloproteinase (MMP)-2 and -9, which has shown efficacy in an animal model of severe traumatic brain injury (TBI). However, SB-3CT is poorly water-soluble and is metabolized primarily to p-hydroxy SB-3CT (2), a more potent inhibitor than SB-3CT. We synthesized the O-phosphate prodrug (3) of compound 2 to enhance its water solubility by more than 2000-fold. The prodrug 3 was a poor MMP inhibitor, but readily hydrolyzed to the active 2 in human blood. Pharmacokinetics and brain distribution studies in mice showed that 2 crossed the blood-brain barrier (BBB) and achieved therapeutic concentrations in the brain. The prodrug 3/compound 2 was evaluated in a mouse model of severe TBI and found to significantly decrease the brain lesion volume and improve neurological outcomes. MMP-9 inhibition by a water-soluble thiirane inhibitor is a promising therapy for treatment of TBI.


Journal of Medicinal Chemistry | 2014

Dual protonophore-chitinase inhibitors dramatically affect O. volvulus molting.

Major Gooyit; Nancy Tricoche; Sara Lustigman; Kim D. Janda

The L3-stage-specific chitinase OvCHT1 has been implicated in the development of Onchocerca volvulus, the causative agent of onchocerciasis. Closantel, a known anthelmintic drug, was previously discovered as a potent and specific OvCHT1 inhibitor. As closantel is also a known protonophore, we performed a simple scaffold modulation to map out the structural features that are relevant for its individual or dual biochemical roles. Furthermore, we present that either OvCHT1 inhibition or protonophoric activity was capable of affecting O. volvulus L3 molting and that the presence of both activities in a single molecule yielded more potent inhibition of the nematode’s developmental process.


Chemical Biology & Drug Design | 2009

Synthesis, kinetic characterization and metabolism of diastereomeric 2-(1-(4-Phenoxyphenylsulfonyl)ethyl)thiiranes as potent gelatinase and MT1-MMP Inhibitors

Major Gooyit; Mijoon Lee; Dusan Hesek; Bill Boggess; Allen G. Oliver; Rafael Fridman; Shahriar Mobashery; Mayland Chang

Gelatinases (MMP‐2 and MMP‐9) have been implicated in a number of pathological conditions, including cancer and cardiovascular disease. Hence, small molecule inhibitors of these enzymes are highly sought for use as potential therapeutic agents. 2‐(4‐Phenoxyphenylsulfonylmethyl)thiirane (SB‐3CT) has previously been demonstrated to be a potent and selective inhibitor of gelatinases, however, it is rapidly metabolized because of oxidation at the para position of the phenoxy ring and at the α‐position to the sulfonyl group. α‐Methyl variants of SB‐3CT were conceived to improve metabolic stability and as mechanistic probes. We describe herein the synthesis and evaluation of these structural variants as potent inhibitors of gelatinases. Two (compounds 5b and 5d) among the four synthetic stereoisomers were found to exhibit slow‐binding inhibition of gelatinases and MMP‐14 (MT1‐MMP), which is a hallmark of the mechanism of this class of inhibitors. The ability of these compounds to inhibit MMP‐2, MMP‐9, and MMP‐14 could target cancer tissues more effectively. Metabolism of the newly synthesized inhibitors showed that both oxidation at the α‐position to the sulfonyl group and oxidation at the para position of the terminal phenyl ring were prevented. Instead oxidation on the thiirane sulfur is the only biotransformation pathway observed for these gelatinase inhibitors.

Collaboration


Dive into the Major Gooyit's collaboration.

Top Co-Authors

Avatar

Mayland Chang

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar

Mark A. Suckow

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar

Kim D. Janda

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Shahriar Mobashery

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mijoon Lee

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar

Zhihong Peng

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar

Bill Boggess

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar

Dusan Hesek

University of Notre Dame

View shared research outputs
Researchain Logo
Decentralizing Knowledge