Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pharmacol Rev ; 74(3): 823-873, 2022 07.
Article in English | MEDLINE | ID: mdl-35738683

ABSTRACT

Carbon monoxide (CO) has been firmly established as an endogenous signaling molecule with a variety of pathophysiological and pharmacological functions, including immunomodulation, organ protection, and circadian clock regulation, among many others. In terms of its molecular mechanism(s) of action, CO is known to bind to a large number of hemoproteins with at least 25 identified targets, including hemoglobin, myoglobin, neuroglobin, cytochrome c oxidase, cytochrome P450, soluble guanylyl cyclase, myeloperoxidase, and some ion channels with dissociation constant values spanning the range of sub-nM to high µM. Although CO's binding affinity with a large number of targets has been extensively studied and firmly established, there is a pressing need to incorporate such binding information into the analysis of CO's biologic response in the context of affinity and dosage. Especially important is to understand the reservoir role of hemoglobin in CO storage, transport, distribution, and transfer. We critically review the literature and inject a sense of quantitative assessment into our analyses of the various relationships among binding affinity, CO concentration, target occupancy level, and anticipated pharmacological actions. We hope that this review presents a picture of the overall landscape of CO's engagement with various targets, stimulates additional research, and helps to move the CO field in the direction of examining individual targets in the context of all of the targets and the concentration of available CO. We believe that such work will help the further understanding of the relationship of CO concentration and its pathophysiological functions and the eventual development of CO-based therapeutics. SIGNIFICANCE STATEMENT: The further development of carbon monoxide (CO) as a therapeutic agent will significantly rely on the understanding of CO's engagement with therapeutically relevant targets of varying affinity. This review critically examines the literature by quantitatively analyzing the intricate relationships among targets, target affinity for CO, CO level, and the affinity state of carboxyhemoglobin and provide a holistic approach to examining the molecular mechanism(s) of action for CO.


Subject(s)
Biological Products , Carbon Monoxide , Carbon Monoxide/metabolism , Carbon Monoxide/pharmacology , Humans , Signal Transduction
2.
J Org Chem ; 89(13): 9551-9556, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38888488

ABSTRACT

Because of endogenous signaling roles of carbon monoxide (CO) and its demonstrated pharmacological effects, there has been extensive interests in developing fluorescent CO probes. Palladium-mediated CO insertion has been successfully used for such applications. However, recent years have seen many publications of using uncatalyzed CO insertion into a hydrazone double bond as a way to sense CO. Such chemistry has no precedents otherwise. Further, the rigor of the CO-sensing work was largely based on using ruthenium-carbonyl complexes such as CORM-3 as CO surrogates, which have been reported to have extensive chemical reactivity and to release largely CO2 instead of CO unless in the presence of a strong nucleophile such as dithionite. For all of these, it is important to reassess the feasibility of such a CO-insertion reaction. By studying two of the reported "CO probes" using CO gas, this study finds no evidence of CO insertion into a hydrazone double bond. Further, the chemical reaction between CO gas and a series of eight hydrazone compounds was conducted, leading to the same conclusion. Such findings are consistent with the state-of-the-art knowledge of carbonylation chemistry and do not support uncatalyzed CO insertion as a mechanism for developing fluorescent CO probes.

3.
Angew Chem Int Ed Engl ; 63(26): e202403880, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38630918

ABSTRACT

Reactive oxygen species (ROS) are critical for cellular signaling. Various pathophysiological conditions are also associated with elevated levels of ROS. Hence, ROS-sensitive triggers have been extensively used for selective payload delivery. Such applications are predicated on two key functions: (1) a sufficient magnitude of concentration difference for the interested ROS between normal tissue/cells and intended sites and (2) appropriate reaction kinetics to ensure a sufficient level of selectivity for payload release. Further, ROS refers to a group of species with varying reactivity, which should not be viewed as a uniform group. In this review, we critically analyze data on the concentrations of different ROS species under various pathophysiological conditions and examine how reaction kinetics affect the success of ROS-sensitive linker chemistry. Further, we discuss different ROS linker chemistry in the context of their applications in drug delivery and imaging. This review brings new insights into research in ROS-triggered delivery, highlights factors to consider in maximizing the chance for success and discusses pitfalls to avoid.


Subject(s)
Reactive Oxygen Species , Reactive Oxygen Species/metabolism , Humans , Drug Delivery Systems , Feasibility Studies , Animals , Kinetics
4.
Med Res Rev ; 43(2): 319-342, 2023 03.
Article in English | MEDLINE | ID: mdl-36177531

ABSTRACT

Nature has the remarkable ability to realize reactions under physiological conditions that normally would require high temperature and other forcing conditions. In doing so, often proximity effects such as simultaneous binding of two reactants in the same pocket and/or strategic positioning of catalytic functional groups are used as ways to achieve otherwise kinetically challenging reactions. Though true biomimicry is challenging, there have been many beautiful examples of how to leverage proximity effects in realizing reactions that otherwise would not readily happen under near-physiological conditions. Along this line, click chemistry is often used to endow proximity effects, and proximity effects are also used to further leverage the facile and bioorthogonal nature of click chemistry. This review brings otherwise seemingly unrelated topics in chemical biology and drug discovery under one unifying theme of mutual leveraging of proximity effects and click chemistry and aims to critically analyze the biomimicry use of such leveraging effects as powerful approaches in chemical biology and drug discovery. We hope that this review demonstrates the power of employing mutual leveraging proximity effects and click chemistry and inspires the development of new strategies that will address unmet needs in chemistry and biology.


Subject(s)
Click Chemistry , Drug Discovery , Humans , Biology
5.
J Am Chem Soc ; 145(1): 78-88, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36548940

ABSTRACT

Extensive studies in the last few decades have led to the establishment of CO as an endogenous signaling molecule and subsequently to the exploration of CO's therapeutic roles. In the current state, there is a critical conundrum in CO-related research: the extensive knowledge of CO's biological effects and yet an insufficient understanding of the quantitative correlations between the CO concentration and biological responses of various natures. This conundrum is partially due to the difficulty in examining precise concentration-response relationships of a gaseous molecule. Another reason is the need for appropriate tools for the sensitive detection and concentration determination of CO in the biological system. We herein report a new chemical approach to the design of fluorescent CO probes through de novo construction of fluorophores by a CO insertion-initiated lactamization reaction, which allows for ultra-low background and exclusivity in CO detection. Two series of CO detection probes have been designed and synthesized using this strategy. Using these probes, we have extensively demonstrated their utility in quantifying CO in blood, tissue, and cell culture and in cellular imaging of CO from exogenous and endogenous sources. The probes described will enable many biology and chemistry labs to study CO's functions in a concentration-dependent fashion with very high sensitivity and selectivity. The chemical and design principles described will also be applicable in designing fluorescent probes for other small molecules.


Subject(s)
Carbon Monoxide , Fluorescent Dyes , Fluorescent Dyes/chemistry
6.
Anal Chem ; 95(23): 9083-9089, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37263968

ABSTRACT

Carbon monoxide (CO) is an endogenous signaling molecule with demonstrated pharmacological effects. For studying CO biology, there is a need for sensitive and selective fluorescent probes for CO as research tools. In developing such probes, CO gas and/or commercially available metal-carbonyl-based "CO-releasing molecules" (CORMs) have been used as CO sources. However, new findings are steadily emerging that some of these commonly used CORMs do not release CO reliably in buffers commonly used for studying such CO probes and have very pronounced chemical reactivities of their own, which could lead to the erroneous identification of "CO probes" that merely detect the CORM used, not CO. This is especially true when the CO-sensing mechanism relies on chemistry that is not firmly established otherwise. Cu2+ can quench the fluorescence of an imine-based fluorophore, DPHP, presumably through complexation. The Cu2+-quenched fluorescence was restored through the addition of CORM-3, a Ru-based CORM. This approach was reported as a new "strategy for detecting carbon monoxide" with the proposed mechanism being dependent on CO reduction of Cu2+ to Cu1+ under near-physiological conditions ( Anal. Chem. 2022, 94, 11298-11306). The study only used CORM-3 as the source of CO. CORM-3 has been reported to have very pronounced redox reactivity and is known not to release CO in an aqueous solution unless in the presence of a strong nucleophile. To assess whether the fluorescent response of the DPHP-Cu(II) cocktail to CORM-3 was truly through detecting CO, we report experiments using both pure CO and CORM-3. We confirm the reported DPHP-Cu(II) response to CORM-3 but not pure CO gas. Further, we did not observe the stated selectivity of DPHP for CO over sulfide species. Along this line, we also found that a reducing agent such as ascorbate was able to induce the same fluorescent turn-on as CORM-3 did. As such, the DPHP-Cu(II) system is not a CO probe and cannot be used to study CO biology. Corollary to this finding, it is critical that future work in developing CO probes uses more than a chemically reactive "CO donor" as the CO source. Especially important will be to confirm the ability of the "CO probe" to detect CO using pure CO gas or another source of CO.

7.
Mol Pharm ; 20(3): 1850-1856, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36802675

ABSTRACT

Carbon monoxide (CO), an endogenous signaling molecule, is known to exert a range of pharmacological effects, including anti-inflammation, organ protection, and antimetastasis in various animal models. We have previously shown the ability of organic prodrugs to deliver CO systemically through oral administration. As part of our efforts for the further development of these prodrugs, we are interested in minimizing the potential negative impact of the "carrier" portion of the prodrug. Along this line, we have previously published our work on using benign "carriers" and physically trapping the "carrier" portion in the gastrointestinal (GI) tract. We herein report our feasibility studies on using immobilized organic CO prodrugs for oral CO delivery while minimizing systemic exposure to the prodrug and the "carrier portion." In doing so, we immobilize a CO prodrug to silica microparticles, which are generally recognized as safe by the US FDA and known to provide large surface areas for loading and water accessibility. The latter point is essential for the hydrophobicity-driven activation of the CO prodrug. Amidation-based conjugation with silica is shown to provide 0.2 mmol/g loading degree, effective prodrug activation in buffer with comparable kinetics as the parent prodrug, and stable tethering to prevent detachment. One representative silica conjugate, SICO-101, is shown to exhibit anti-inflammation activity in LPS-challenged RAW264.7 cells and to deliver CO systemically in mice through oral administration and GI CO release. We envision this strategy as a general approach for oral CO delivery to treat systemic and GI-specific inflammatory conditions.


Subject(s)
Prodrugs , Mice , Animals , Prodrugs/pharmacology , Feasibility Studies , Carbon Monoxide , Anti-Inflammatory Agents/pharmacology , Gastrointestinal Tract , Excipients
8.
J Enzyme Inhib Med Chem ; 38(1): 2276663, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37955285

ABSTRACT

Conjugation of drugs with biotin is a widely studied strategy for targeted drug delivery. The structure-activity relationship (SAR) studies through H3-biotin competition experiments conclude with the presence of a free carboxylic acid being essential for its uptake via the sodium-dependent multivitamin transporter (SMVT, the major biotin transporter). However, biotin conjugation with a payload requires modification of the carboxylic acid to an amide or ester group. Then, there is the question as to how/whether the uptake of biotin conjugates goes through the SMVT. If not, then what is the mechanism? Herein, we present known uptake mechanisms of biotin and its applications reported in the literature. We also critically analyse possible uptake mechanism(s) of biotin conjugates to address the disconnect between the results from SMVT-based SAR and "biotin-facilitated" targeted drug delivery. We believe understanding the uptake mechanism of biotin conjugates is critical for their future applications and further development.


Subject(s)
Biotin , Symporters , Drug Delivery Systems , Carboxylic Acids
9.
Chem Rev ; 120(24): 13273-13311, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33089988

ABSTRACT

Nature is full of examples of symbiotic relationships. The critical symbiotic relation between host and mutualistic bacteria is attracting increasing attention to the degree that the gut microbiome is proposed by some as a new organ system. The microbiome exerts its systemic effect through a diverse range of metabolites, which include gaseous molecules such as H2, CO2, NH3, CH4, NO, H2S, and CO. In turn, the human host can influence the microbiome through these gaseous molecules as well in a reciprocal manner. Among these gaseous molecules, NO, H2S, and CO occupy a special place because of their widely known physiological functions in the host and their overlap and similarity in both targets and functions. The roles that NO and H2S play have been extensively examined by others. Herein, the roles of CO in host-gut microbiome communication are examined through a discussion of (1) host production and function of CO, (2) available CO donors as research tools, (3) CO production from diet and bacterial sources, (4) effect of CO on bacteria including CO sensing, and (5) gut microbiome production of CO. There is a large amount of literature suggesting the "messenger" role of CO in host-gut microbiome communication. However, much more work is needed to begin achieving a systematic understanding of this issue.


Subject(s)
Bacteria/metabolism , Carbon Monoxide/metabolism , Gastrointestinal Microbiome/physiology , Animals , Bacterial Physiological Phenomena , Humans , Symbiosis
10.
J Enzyme Inhib Med Chem ; 37(1): 2370-2381, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36043494

ABSTRACT

Overexpression of ubiquitin ligase MDM2 causes depletion of the p53 tumour-suppressor and thus leads to cancer progression. In recent years, anthraquinone analogs have received significant attention due to their ability to downregulate MDM2, thereby promoting p53-induced apoptosis. Previously, we have developed potent anthraquinone compounds having the ability to upregulate p53 via inhibition of MDM2 in both cell culture and animal models of acute lymphocytic leukaemia. Earlier work was focussed on mechanistic work, pharmacological validation of this class of compounds in animal models, and mapping out structural space that allows for further modification and optimisation. Herein, we describe our work in optimising the substituents on the two phenol hydroxyl groups. It was found that the introduction of an alkylketone moiety led to a potent series of analogs with BW-AQ-350 being the most potent compound yet (IC50 = 0.19 ± 0.01 µM) which exerts cytotoxicity by inducing MDM2 degradation and p53 upregulation.


Subject(s)
Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53 , Animals , Anthraquinones/pharmacology , Apoptosis , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation
11.
Anal Chem ; 93(12): 5317-5326, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33745269

ABSTRACT

Ruthenium-based CO-releasing molecules (CO-RMs), CORM-2 and CORM-3, have been widely used as surrogates of CO for studying its biological effects in vitro and in vivo with much success. However, several previous solution-phase and in vitro studies have revealed the ability of such CO-RMs to chemically modify proteins and reduce aromatic nitro groups due to their intrinsic chemical reactivity under certain conditions. In our own work of studying the cytoprotective effects of CO donors, we were in need of assessing chemical factors that could impact the interpretation of results from CO donors including CORM-2,3 in various in vitro assays. For this, we examined the effects of CORM-2,3 toward representative reagents commonly used in various bioassays including resazurin, tetrazolium salts, nitrites, and azide-based H2S probes. We have also examined the effect of CORM-2,3 on glutathione disulfide (GSSG), which is a very important redox regulator. Our studies show the ability of these CO-RMs to induce a number of chemical and/or spectroscopic changes for several commonly used biological reagents under near-physiological conditions. These reactions/spectroscopic changes cannot be duplicated with CO-deleted CO-RMs (iCORMs), which are often used as negative controls. Furthermore, both CORM-2 and -3 are capable of consuming and reducing GSSG in solution. We hope that the results described will help in the future design of control experiments using Ru-based CO-RMs.


Subject(s)
Organometallic Compounds , Ruthenium , Carbon Monoxide , Indicators and Reagents , Nitrites
12.
Bioorg Med Chem ; 44: 116297, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34243045

ABSTRACT

Delivering a therapeutically active gaseous molecule represents very unique challenges in terms of both precise dosing and concentration assessment. To overcome these obstacles, there have been recent reports of using prodrug approaches for the in-vitro and in-vivo generation of carbon monoxide (CO), which is an endogenous signaling molecule with validated therapeutic efficacy in a range of animal models. Some key components of these approaches include the use of a hydrophobicity-driven Diels-Alder reaction under physiological conditions followed by a cheletropic reaction of the corresponding norbornadien-7-one intermediate, leading to extrusion of CO. With proper design, the same approach also leads to the formation of a fluorescent reporter, allowing for quantitative assessment of the amount of CO released. All these allow for a strategy of "click, release, and fluoresce" in delivering a precise dose of carbon monoxide with the ability to "self-report" delivery quantity and efficiency. This strategy has also been further refined to construct a CO delivery platform with additional functionalities such as bioorthogonal labeling, targeting, triggered release, and simultaneously delivery of more than one payload. This review highlights recent developments in this area.


Subject(s)
Carbon Monoxide/chemistry , Fluorescence , Fluorescent Dyes/chemical synthesis , Click Chemistry , Fluorescent Dyes/chemistry , Molecular Structure
13.
Proc Natl Acad Sci U S A ; 115(10): E2302-E2310, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463714

ABSTRACT

Ischemia reperfusion injury (IRI) is the predominant tissue insult associated with organ transplantation. Treatment with carbon monoxide (CO) modulates the innate immune response associated with IRI and accelerates tissue recovery. The mechanism has been primarily descriptive and ascribed to the ability of CO to influence inflammation, cell death, and repair. In a model of bilateral kidney IRI in mice, we elucidate an intricate relationship between CO and purinergic signaling involving increased CD39 ectonucleotidase expression, decreased expression of Adora1, with concomitant increased expression of Adora2a/2b. This response is linked to a >20-fold increase in expression of the circadian rhythm protein Period 2 (Per2) and a fivefold increase in serum erythropoietin (EPO), both of which contribute to abrogation of kidney IRI. CO is ineffective against IRI in Cd39-/- and Per2-/- mice or in the presence of a neutralizing antibody to EPO. Collectively, these data elucidate a cellular signaling mechanism whereby CO modulates purinergic responses and circadian rhythm to protect against injury. Moreover, these effects involve CD39- and adenosinergic-dependent stabilization of Per2. As CO also increases serum EPO levels in human volunteers, these findings continue to support therapeutic use of CO to treat IRI in association with organ transplantation, stroke, and myocardial infarction.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Carbon Monoxide/administration & dosage , Kidney Diseases/drug therapy , Kidney/drug effects , Period Circadian Proteins/metabolism , Reperfusion Injury/prevention & control , Animals , Antigens, CD/genetics , Apyrase/genetics , Disease Models, Animal , Humans , Kidney/blood supply , Kidney/metabolism , Kidney/physiopathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Male , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Reperfusion Injury/genetics , Reperfusion Injury/metabolism
14.
Int J Mol Sci ; 22(10)2021 May 14.
Article in English | MEDLINE | ID: mdl-34069086

ABSTRACT

Hydrogen sulfide (H2S) is an endogenously produced molecule with anti-inflammatory and cytoprotective properties. We aimed to investigate for the first time if a novel, esterase-sensitive H2S-prodrug, BW-HS-101 with the ability to release H2S in a controllable manner, prevents gastric mucosa against acetylsalicylic acid-induced gastropathy on microscopic and molecular levels. Wistar rats were pretreated intragastrically with vehicle, BW-HS-101 (0.5-50 µmol/kg) or its analogue without the ability to release H2S, BW-iHS-101 prior to ASA administration (125 mg/kg, intragastrically). BW-HS-101 was administered alone or in combination with nitroarginine (L-NNA, 20 mg/kg, intraperitoneally) or zinc protoporphyrin IX (10 mg/kg, intraperitoneally). Gastroprotective effects of BW-HS-101 were additionally evaluated against necrotic damage induced by intragastrical administration of 75% ethanol. Gastric mucosal damage was assessed microscopically, and gastric blood flow was determined by laser flowmetry. Gastric mucosal DNA oxidation and PGE2 concentration were assessed by ELISA. Serum and/or gastric protein concentrations of IL-1α, IL-1ß, IL-2, IL-4, IL-6, IL-10, IL-13, VEGF, GM-CSF, IFN-γ, TNF-α, and EGF were determined by a microbeads/fluorescent-based multiplex assay. Changes in gastric mucosal iNOS, HMOX-1, SOCS3, IL1-R1, IL1-R2, TNF-R2, COX-1, and COX-2 mRNA were assessed by real-time PCR. BW-HS-101 or BW-iHS-101 applied at a dose of 50 µmol/kg protected gastric mucosa against ASA-induced gastric damage and prevented a decrease in the gastric blood flow level. H2S prodrug decreased DNA oxidation, systemic and gastric mucosal inflammation with accompanied upregulation of SOCS3, and EGF and HMOX-1 expression. Pharmacological inhibition of nitric oxide (NO) synthase but not carbon monoxide (CO)/heme oxygenase (HMOX) activity by L-NNA or ZnPP, respectively, reversed the gastroprotective effect of BW-HS-101. BW-HS-101 also protected against ethanol-induced gastric injury formation. We conclude that BW-HS-101, due to its ability to release H2S in a controllable manner, prevents gastric mucosa against drugs-induced gastropathy, inflammation and DNA oxidation, and upregulate gastric microcirculation. Gastroprotective effects of this H2S prodrug involves endogenous NO but not CO activity and could be mediated by cytoprotective and anti-inflammatory SOCS3 and EGF pathways.


Subject(s)
Gastric Mucosa/drug effects , Hydrogen Sulfide/pharmacokinetics , Protective Agents/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Aspirin/adverse effects , DNA/metabolism , Drug Liberation , Ethanol/toxicity , Gastric Mucosa/blood supply , Gastric Mucosa/pathology , Gastritis/chemically induced , Gastritis/drug therapy , Gastritis/pathology , Gene Expression Regulation/drug effects , Male , Nitric Oxide/metabolism , Nitroarginine/administration & dosage , Nitroarginine/pharmacology , Prodrugs/pharmacokinetics , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , Protective Agents/administration & dosage , Protoporphyrins/administration & dosage , Protoporphyrins/pharmacology , Rats, Wistar
15.
Med Res Rev ; 40(4): 1147-1177, 2020 07.
Article in English | MEDLINE | ID: mdl-31820474

ABSTRACT

Treating acute kidney injury (AKI) represents an important unmet medical need both in terms of the seriousness of this medical problem and the number of patients. There is also a large untapped market opportunity in treating AKI. Over the years, there has been much effort in search of therapeutics with minimal success. However, over the same time period, new understanding of the underlying pathobiology and molecular mechanisms of kidney injury have undoubtedly helped the search for new therapeutics. Along this line, carbon monoxide (CO) has emerged as a promising therapeutic agent because of its demonstrated cytoprotective, and immunomodulatory effects. CO has also been shown to sensitize cancer, but not normal cells, to chemotherapy. This is particularly important in treating cisplatin-induced AKI, a common clinical problem that develops in patients receiving cisplatin therapies for a number of different solid organ malignancies. This review will examine and make the case that CO be developed into a therapeutic agent against AKI.


Subject(s)
Acute Kidney Injury/drug therapy , Carbon Monoxide/therapeutic use , Acute Kidney Injury/etiology , Animals , Carbon Monoxide/administration & dosage , Clinical Trials as Topic , Disease Models, Animal , Drug Delivery Systems , Humans
16.
Med Res Rev ; 40(6): 2682-2713, 2020 11.
Article in English | MEDLINE | ID: mdl-32803765

ABSTRACT

Smart drugs, such as antibody-drug conjugates, for targeted therapy rely on the ability to deliver a warhead to the desired location and to achieve activation at the same site. Thus, designing a smart drug often requires proper linker chemistry for tethering the warhead with a vehicle in such a way that either allows the active drug to retain its potency while being tethered or ensures release and thus activation at the desired location. Recent years have seen much progress in the design of new linker activation strategies. Herein, we review the recent development of chemical strategies used to link the warhead with a delivery vehicle for preferential cleavage at the desired sites.


Subject(s)
Pharmaceutical Preparations , Drug Delivery Systems , Humans
17.
Biochem Biophys Res Commun ; 524(3): 730-735, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32035620

ABSTRACT

Post-translational modifications (PTMs) play pivotal roles in controlling the stability and activity of the tumor suppressor p53 in response to distinct stressors. Here we report an unexpected finding of a short chain fatty acid modification of p53 in human cells. Crotonic acid (CA) treatment induces p53 crotonylation, but surprisingly reduces its protein, but not mRNA level, leading to inhibition of p53 activity in a dose dependent fashion. Surprisingly this crotonylation targets serine 46, instead of any predicted lysine residues, of p53, as detected in TCEP-probe labeled crotonylation and anti-crotonylated peptide antibody reaction assays. This is further confirmed by substitution of serine 46 with alanine, which abolishes p53 crotonylation in vitro and in cells. CA increases p53-dependent glycolytic activity, and augments cancer cell proliferation in response to metabolic or DNA damage stress. Since serine 46 is only found in human p53, our studies unveil an unconventional PTM unique for human p53, impairing its activity in response to CA. Because CA is likely produced by the gut microbiome, our results also predict that this type of PTM might play a role in early human colorectal neoplasia development by negating p53 activity without mutation of this tumor suppressor gene.


Subject(s)
Crotonates/metabolism , Protein Processing, Post-Translational , Serine/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cell Proliferation , Crotonates/chemistry , Glucose/deficiency , Glycolysis , Humans , Lysine/metabolism , Mitochondria/metabolism , Tumor Suppressor Protein p53/chemistry
18.
Bioorg Med Chem Lett ; 30(2): 126786, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31753697

ABSTRACT

Previously, we reported a class of MDM2-MDM4 dimerization inhibitors that upregulate p53 and showed potent anticancer activity in animal models. However, water solubility hinders their further development. Herein we describe our effort to develop a prodrug approach that overcomes the solubility problem. The prodrug of BW-AQ-238, a potent anthraquinone analog, was made by esterification of the hydroxyl group with various natural amino acids. Cytotoxicity of these compounds toward Hela and EU-1 cells, their aqueous solubility, and the release kinetics of these prodrugs in buffer and in the presence of hydrolytic enzymes were studied. The results demonstrate that the amino acid prodrug approach significantly improved the water solubility while maintaining the potency of the parent drug.


Subject(s)
Amino Acids/chemistry , Anthraquinones/chemistry , Prodrugs/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Anthraquinones/metabolism , Anthraquinones/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Half-Life , Humans , Prodrugs/metabolism , Prodrugs/pharmacology , Structure-Activity Relationship , Up-Regulation/drug effects
19.
Chem Soc Rev ; 48(4): 1077-1094, 2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30724944

ABSTRACT

Prodrug approaches represent an excellent solution to certain pharmaceutical issues commonly encountered in the drug discovery and development process. Along this line, the chemistry needed for the bio-reversible derivatization of drug functional groups for on-demand release is critical. In recent years, "click and release" approaches have shown great promise in the design of prodrugs because of their bioorthogonality and controlled bond-cleavage, which help ensure prodrug stability during circulation and ready cleavage at the desired site of action. This review highlights recent developments of this research field and discusses issues yet to be addressed.


Subject(s)
Click Chemistry/methods , Drug Delivery Systems/methods , Prodrugs/chemistry , Animals , Delayed-Action Preparations/chemistry , Drug Liberation , Humans , Molecular Structure , Structure-Activity Relationship
20.
Med Chem Res ; 29(7): 1199-1210, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32719577

ABSTRACT

We previously reported a series of p53-elevating anthraquinone compounds with considerable cytotoxicity for acute lymphatic leukemia (ALL) cells. To further develop this class of compounds, we examined the effect of a few key structural features on the anticancer structure-activity relationship in ALL cells. The active analogs showed comparable cytotoxicity and upregulation of p53 but did not induce significant downregulation of MDM2 as seen with the lead compound AQ-101, indicating the importance of the anthraquinone core scaffold for MDM2 regulation. The result from the current study not only contributes to the SAR framework of these anthraquinone derivatives but also opens up new chemical space for further optimization work.

SELECTION OF CITATIONS
SEARCH DETAIL