Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38802678

ABSTRACT

Lewisite, a chemical warfare agent, causes skin blisters, erythema, edema, and inflammation, requiring mitigation strategies in case of accidental or deliberate exposure. 4-phenyl butyric acid (4-PBA), a chemical chaperone, reduces endoplasmic reticulum stress and skin inflammation. The study aimed to encapsulate 4-PBA in microsponges for effective, sustained delivery against lewisite injury. Porous microsponges in a topical gel would potentially sustain delivery and improve residence time on the skin. Microsponges were developed using the quasi-emulsion solvent diffusion method with Eudragit RS100. Optimized formulation showed 10.58%w/w drug loading was incorporated in a carboxymethylcellulose (CMC) and Carbopol gel for in vitro release and permeation testing using dermatomed human skin. A sustained release was obtained from all vehicles in the release study, and IVPT results showed that compared to the control (41.52 ± 2.54 µg/sq.cm), a sustained permeation profile with a reduced delivery was observed for microsponges in PBS (14.16 ± 1.23 µg/sq.cm) along with Carbopol 980 gel (12.55 ± 1.41 µg/sq.cm), and CMC gel (10.09 ± 1.23 µg/sq.cm) at 24 h. Optimized formulation showed significant protection against lewisite surrogate phenyl arsine oxide (PAO) challenged skin injury in Ptch1+/-/SKH-1 hairless mice at gross and molecular levels. A reduction in Draize score by 29%, a reduction in skin bifold thickness by 8%, a significant reduction in levels of IL-1ß, IL6, and GM-CSF by 54%, 30%, and 55%, respectively, and a reduction in apoptosis by 31% was observed. Thus, the translational feasibility of 4-PBA microsponges for effective, sustained delivery against lewisite skin injury is demonstrated.

2.
J Pharmacol Exp Ther ; 388(2): 605-612, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37699712

ABSTRACT

Arsenicals are deadly chemical warfare agents that primarily cause death through systemic capillary fluid leakage and hypovolemic shock. Arsenical exposure is also known to cause acute kidney injury, a condition that contributes to arsenical-associated death due to the necessity of the kidney in maintaining whole-body fluid homeostasis. Because of the global health risk that arsenicals pose, a nuanced understanding of how arsenical exposure can lead to kidney injury is needed. We used a nontargeted transcriptional approach to evaluate the effects of cutaneous exposure to phenylarsine oxide, a common arsenical, in a murine model. Here we identified an upregulation of metabolic pathways such as fatty acid oxidation, fatty acid biosynthesis, and peroxisome proliferator-activated receptor (PPAR)-α signaling in proximal tubule epithelial cell and endothelial cell clusters. We also revealed highly upregulated genes such as Zbtb16, Cyp4a14, and Pdk4, which are involved in metabolism and metabolic switching and may serve as future therapeutic targets. The ability of arsenicals to inhibit enzymes such as pyruvate dehydrogenase has been previously described in vitro. This, along with our own data, led us to conclude that arsenical-induced acute kidney injury may be due to a metabolic impairment in proximal tubule and endothelial cells and that ameliorating these metabolic effects may lead to the development of life-saving therapies. SIGNIFICANCE STATEMENT: In this study, we demonstrate that cutaneous arsenical exposure leads to a transcriptional shift enhancing fatty acid metabolism in kidney cells, indicating that metabolic alterations might mechanistically link topical arsenical exposure to acute kidney injury. Targeting metabolic pathways may generate promising novel therapeutic approaches in combating arsenical-induced acute kidney injury.


Subject(s)
Acute Kidney Injury , Arsenicals , Mice , Humans , Animals , Endothelial Cells/metabolism , Kidney/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Epithelial Cells/metabolism , Fatty Acids/metabolism , Arsenicals/adverse effects , Arsenicals/metabolism
3.
Int J Pharm ; 647: 123547, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-37884214

ABSTRACT

Lewisite is a chemical warfare agent intended for use in World War and a potential threat to the civilian population due to presence in stockpiles or accidental exposure. Lewisite-mediated skin injury is characterized by acute erythema, pain, and blister formation. N-acetyl cysteine (NAC) is an FDA-approved drug for acetaminophen toxicity, identified as a potential antidote against lewisite. In the present study, we have explored the feasibility of rapid NAC delivery through transdermal route for potentially treating chemical warfare toxicity. NAC is a small, hydrophilic molecule with limited passive delivery through the skin. Using skin microporation with dissolving microneedles significantly enhanced the delivery of NAC into and across dermatomed human skin in our studies. Microporation followed by application of solution (poke-and-solution) resulted in the highest in vitro delivery (509.84 ± 155.04 µg/sq·cm) as compared to poke-and-gel approach (474.91 ± 70.09 µg/sq·cm) and drug-loaded microneedles (226.89 ± 33.41 µg/sq·cm). The lag time for NAC delivery through poke-and-solution approach (0.23 ± 0.04 h) was close to gel application (0.25 ± 0.02 h), with the highest for drug-loaded microneedles (1.27 ± 1.16 h). Thus, we successfully demonstrated the feasibility of rapid NAC delivery using various skin microporation approaches for potential treatment against lewisite-mediated skin toxicity.


Subject(s)
Acetylcysteine , Antidotes , Humans , Administration, Cutaneous , Skin , Drug Delivery Systems , Needles
4.
Ann N Y Acad Sci ; 1515(1): 168-183, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35678766

ABSTRACT

Arsenical vesicants cause skin inflammation, blistering, and pain. The lack of appropriate animal models causes difficulty in defining their molecular pathogenesis. Here, Ptch1+/- /C57BL/6 mice were employed to investigate the pathobiology of the arsenicals lewisite and phenylarsine oxide (PAO). Following lewisite or PAO challenge (24 h), the skin of animals becomes grayish-white, thick, leathery, and wrinkled with increased bi-fold thickness, Draize score, and necrotic patches. In histopathology, infiltrating leukocytes (macrophages and neutrophils), epidermal-dermal separation, edema, apoptotic cells, and disruption of tight and adherens junction proteins can be visualized. PCR arrays and nanoString analyses showed significant increases in cytokines/chemokines and other proinflammatory mediators. As hair follicles (HFs), which provide an immune-privileged environment, may affect immune cell trafficking and consequent inflammatory responses, we compared the pathogenesis of these chemicals in this model to that in Ptch1+/- /SKH-1 hairless mice. Ptch1+/- /SKH-1 mice have rudimentary, whereas Ptch1+/- /C57BL/6 mice have well-developed HFs. Although no significant differences were observed in qualitative inflammatory responses between the two strains, levels of cytokines/chemokines differed. Importantly, the mechanism of inflammation was identical; both reactive oxygen species induction and consequent activation of unfolded protein response signaling were similar. These data reveal that the acute molecular pathogenesis of arsenicals in these two murine models is similar.


Subject(s)
Arsenicals , Chemical Warfare Agents , Animals , Chemical Warfare Agents/metabolism , Chemokines , Cytokines/metabolism , Hair Follicle/metabolism , Hair Follicle/pathology , Inflammation/pathology , Irritants , Mice , Mice, Hairless , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Skin/metabolism
5.
Mol Carcinog ; 61(8): 737-751, 2022 08.
Article in English | MEDLINE | ID: mdl-35472745

ABSTRACT

Aberrant activation of multiple complex signaling pathways underlies the pathogenesis of rhabdomyosarcoma (RMS), which remains a cause of mortality in approximately 30% of children with RMS. Bromodomain and extraterminal (BET) domain chromatin remodeling regulates several of these pathways. Here, we targeted bromodomain 4 (BRD4) in combination with another molecular metabolic tumor driver, the Akt/mTOR signaling pathway, to provide a highly effective treatment for this neoplasm. We demonstrated that a nexus of these two molecular pathways underlies RMS pathogenesis. Our data show that the combined inhibition of the BET bromodomain and mTORC1/2 signaling abrogates aggressive RMS growth. Thus, the bromodomain inhibitor RVX-208 significantly augmented the therapeutic effects of the dual mTORC1/2 inhibitors, OSI-027 and PP242, both in vitro and in a human xenograft murine model. Drug-treated residual tumors showed a decrease in the activation of underlying signaling mechanisms characterized by a reduction in the expression of p-AKT, p-mTOR, p-p70S6K, cyclin D1, and proliferation. Our ChIP-seq data demonstrated that RVX-208 effectively blocked BRD4 occupancy on its target promoters. ChIP-qPCR assays further confirmed that RVX-208 treatment resulted in a significant decrease in H3K27ac and H4K8ac signals at their target loci. While single RVX-208 treatment induces apoptosis and a single mTORC1/2 inhibitor induces macropinocytosis, their combined treatment led to necroptosis-mediated cell death. These data suggest that combined treatment with drugs targeting BRD4 and mTORC1/2 may be an effective therapeutic intervention for drug-resistant RMS.


Subject(s)
Nuclear Proteins , Rhabdomyosarcoma , Animals , Apoptosis , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Child , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 64: 128696, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35318165

ABSTRACT

Arsenicals belong to the class of chemical warfare agents known as vesicants, which are highly reactive, toxic and cause robust inflammatory response. Cutaneous exposure to arsenicals causes a wide range of systemic organ damage, beginning with cutaneous injuries, and later manifest multi-organ damage and death. Thus, the development of suitable antidotes that can effectively block injury following exposure to these agents is of great importance. Bromodomain 4 (BRD4), a member of the bromodomain and extra terminal domain (BET) family, plays crucial role in regulating transcription of inflammatory, proliferation and cell cycle genes. In this context, the development of potent small molecule inhibitors of BRD4 could serve as potential antidotes for arsenicals. Herein, we describe the synthesis and biological evaluation of a series of compounds.


Subject(s)
Arsenicals , Anti-Inflammatory Agents/chemistry , Antidotes/pharmacology , Arsenicals/pharmacology , Arsenicals/therapeutic use , Nuclear Proteins/metabolism , Transcription Factors/metabolism
7.
Ann N Y Acad Sci ; 1480(1): 155-169, 2020 11.
Article in English | MEDLINE | ID: mdl-32885420

ABSTRACT

Lewisite and many other similar arsenicals are warfare vesicants developed and weaponized for use in World Wars I and II. These chemicals, when exposed to the skin and other epithelial tissues, cause rapid severe inflammation and systemic damage. Here, we show that topically applied arsenicals in a murine model produce significant acute kidney injury (AKI), as determined by an increase in the AKI biomarkers NGAL and KIM-1. An increase in reactive oxygen species and ER stress proteins, such as ATF4 and CHOP, correlated with the induction of these AKI biomarkers. Also, TUNEL staining of CHOP-positive renal tubular cells suggests CHOP mediates apoptosis in these cells. A systemic inflammatory response characterized by a significant elevation in inflammatory mediators, such as IL-6, IFN-α, and COX-2, in the kidney could be the underlying cause of AKI. The mechanism of arsenical-mediated inflammation involves activation of AMPK/Nrf2 signaling pathways, which regulate heme oxygenase-1 (HO-1). Indeed, HO-1 induction with cobalt protoporphyrin (CoPP) treatment in arsenical-treated HEK293 cells afforded cytoprotection by attenuating CHOP-associated apoptosis and cytokine mRNA levels. These results demonstrate that topical exposure to arsenicals causes AKI and that HO-1 activation may serve a protective role in this setting.


Subject(s)
Acute Kidney Injury , Apoptosis/drug effects , Arsenicals , Chemical Warfare Agents/poisoning , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Activating Transcription Factor 4/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Biomarkers/metabolism , Cyclooxygenase 2/metabolism , Enzyme Activation/drug effects , HEK293 Cells , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Interleukin-6/metabolism , Mice , Mice, Hairless , NF-E2-Related Factor 2/metabolism , Transcription Factor CHOP/metabolism
8.
Article in English | MEDLINE | ID: mdl-32684991

ABSTRACT

This study evaluated the topical delivery of nordihydroguaretic acid (NDGA), a molecule that can potentially alleviate cutaneous damage caused by exposure to arsenic warfare chemicals. N-acetylcysteine (NAC 0.2% w/v) was added as an antioxidant, preventing the oxidation of NDGA to toxic quinones. A 24 h study was performed to arrive at a minimum concentration of NDGA needed to deliver maximum drug. A solution of 3% w/v delivered the maximum amount of drug at the end of 24 h (37.45 ± 4.32 µg). Short duration studies were carried out to determine the time needed to saturate skin with NDGA. There was no significant difference in the skin concentrations for 24 h and 8 h (14.89 ± 2.36 µg), due to skin saturation. However, there was significant difference in the amount of drug delivered to the epidermis (12.29 ± 1.87 µg) and dermis (2.54 ± 0.56 µg) at the end of 8 h. Solution of NDGA was applied on UV treated skin to assess changes in drug delivery. In vivo studies revealed that 3% NDGA was non-toxic for topical administration.

9.
Proc Natl Acad Sci U S A ; 116(49): 24583-24592, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31732667

ABSTRACT

The mammalian target of rapamycin (mTOR) pathway, which plays a critical role in regulating cellular growth and metabolism, is aberrantly regulated in the pathogenesis of a variety of neoplasms. Here we demonstrate that dual mTORC1/mTORC2 inhibitors OSI-027 and PP242 cause catastrophic macropinocytosis in rhabdomyosarcoma (RMS) cells and cancers of the skin, breast, lung, and cervix, whereas the effects are much less pronounced in immortalized human keratinocytes. Using RMS as a model, we characterize in detail the mechanism of macropinocytosis induction. Macropinosomes are distinct from endocytic vesicles and autophagosomes in that they are single-membrane bound vacuoles formed by projection, ruffling, and contraction of plasma membranes. They are positive for EEA-1 and LAMP-1 and contain watery fluid but not organelles. The vacuoles then merge and rupture, killing the cells. We confirmed the inhibition of mTORC1/mTORC2 as the underpinning mechanism for macropinocytosis. Exposure to rapamycin, an mTORC1 inhibitor, or mTORC2 knockdown alone had little or reduced effect relative to the combination. We further demonstrate that macropinocytosis depends on MKK4 activated by elevated reactive oxygen species. In a murine xenograft model, OSI-027 reduced RMS tumor growth. Molecular characterization of the residual tumors was consistent with the induction of macropinocytosis. Furthermore, relative to the control xenograft tumors, the residual tumors manifested reduced expression of cell proliferation markers and proteins that drive the epithelial mesenchymal transition. These data indicate a role of mTORC2 in regulating tumor growth by macropinocytosis and suggest that dual inhibitors could help block refractory or recurrent RMS and perhaps other neoplasms and other cancer as well.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Pinocytosis/drug effects , Purines/pharmacology , Triazines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Imidazoles/administration & dosage , MAP Kinase Kinase 4/metabolism , Mice, Nude , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Triazines/administration & dosage , Vacuoles/drug effects , Vacuoles/pathology , Xenograft Model Antitumor Assays
10.
Mol Neurobiol ; 56(9): 6663, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31264091

ABSTRACT

The original version of this article unfortunately contained a mistake. The acknowledgment published was incomplete.

11.
Am J Physiol Renal Physiol ; 314(6): F1166-F1176, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29361668

ABSTRACT

Lewisite (2-chlorovinyldichloroarsine) is an organic arsenical chemical warfare agent that was developed and weaponized during World Wars I/II. Stockpiles of lewisite still exist in many parts of the world and pose potential environmental and human health threat. Exposure to lewisite and similar chemicals causes intense cutaneous inflammatory response. However, morbidity and mortality in the exposed population is not only the result of cutaneous damage but is also a result of systemic injury. Here, we provide data delineating the pathogenesis of acute kidney injury (AKI) following cutaneous exposure to lewisite and its analog phenylarsine oxide (PAO) in a murine model. Both agents caused renal tubular injury, characterized by loss of brush border in proximal tubules and tubular cell apoptosis accompanied by increases in serum creatinine, neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1. Interestingly, lewisite exposure enhanced production of reactive oxygen species (ROS) in the kidney and resulted in the activation of autophagic and DNA damage response (DDR) signaling pathways with increased expression of beclin-1, autophagy-related gene 7, and LC-3A/B-II and increased phosphorylation of γ-H2A.X and checkpoint kinase 1/2, respectively. Terminal deoxyribonucleotide-transferase-mediated dUTP nick-end labeling-positive cells were detected in renal tubules along with enhanced proapoptotic BAX/cleaved caspase-3 and reduced antiapoptotic BCL2. Scavenging ROS by cutaneous postexposure application of the antioxidant N-acetyl-l-cysteine reduced lewisite-induced autophagy and DNA damage. In summary, we provide evidence that topical exposure to lewisite causes AKI. The molecular mechanism underlying these changes involves ROS-dependent activation of autophagy and DDR pathway associated with the induction of apoptosis.


Subject(s)
Acute Kidney Injury/chemically induced , Arsenicals/adverse effects , Autophagy , Chemical Warfare Agents/adverse effects , DNA Damage , Kidney/pathology , Skin Absorption , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Arsenicals/metabolism , Chemical Warfare Agents/metabolism , Cytokines/metabolism , Female , HEK293 Cells , Humans , Kidney/metabolism , Male , Mice, Hairless , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
12.
Article in English | MEDLINE | ID: mdl-28756121

ABSTRACT

BACKGROUND: Long-term survival of lung transplant recipients (LTRs) is limited by the occurrence of bronchiolitis obliterans syndrome (BOS). Recent evidence suggests a role for microbiome alterations in the occurrence of BOS, although the precise mechanisms are unclear. In this study we evaluated the relationship between the airway microbiome and distinct subsets of immunoregulatory myeloid-derived suppressor cells (MDSCs) in LTRs. METHODS: Bronchoalveolar lavage (BAL) and simultaneous oral wash and nasal swab samples were collected from adult LTRs. Microbial genomic DNA was isolated, 16S rRNA genes amplified using V4 primers, and polymerase chain reaction (PCR) products sequenced and analyzed. BAL MDSC subsets were enumerated using flow cytometry. RESULTS: The oral microbiome signature differs from that of the nasal, proximal and distal airway microbiomes, whereas the nasal microbiome is closer to the airway microbiome. Proximal and distal airway microbiome signatures of individual subjects are distinct. We identified phenotypic subsets of MDSCs in BAL, with a higher proportion of immunosuppressive MDSCs in the proximal airways, in contrast to a preponderance of pro-inflammatory MDSCs in distal airways. Relative abundance of distinct bacterial phyla in proximal and distal airways correlated with particular airway MDSCs. Expression of CCAAT/enhancer binding protein (C/EBP)-homologous protein (CHOP), an endoplasmic (ER) stress sensor, was increased in immunosuppressive MDSCs when compared with pro-inflammatory MDSCs. CONCLUSIONS: The nasal microbiome closely resembles the microbiome of the proximal and distal airways in LTRs. The association of distinct microbial communities with airway MDSCs suggests a functional relationship between the local microbiome and MDSC phenotype, which may contribute to the pathogenesis of BOS.

13.
Sci Rep ; 6: 34865, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27725709

ABSTRACT

Arsenicals are painful, inflammatory and blistering causing agents developed as chemical weapons in World War I/II. However, their large stockpiles still exist posing threat to public health. Phenylarsine oxide (PAO), a strong oxidant and a prototype arsenical is tested for its suitability to defining molecular mechanisms underlying arsenicals-mediated tissue injury. Topically applied PAO induces cutaneous erythema, edema and micro-blisters. These gross inflammatory responses were accompanied by the enhanced production of pro-inflammatory cytokines, ROS and unfolded protein response (UPR) signaling activation. To demonstrate the involvement of UPR in the pathobiology of these lesions, we employed chemical chaperone, 4-phenylbutyric acid (4-PBA) which attenuates UPR. 4-PBA significantly reduced PAO-induced inflammation and blistering. Similar to its effects in murine epidermis, a dose- and time-dependent upregulation of ROS, cytokines, UPR proteins (GRP78, p-PERK, p-eIF2α, ATF4 and CHOP) and apoptosis were observed in PAO-treated human skin keratinocytes NHEK and HaCaT. In addition, 4-PBA significantly restored these molecular alterations in these cells. Employing RNA interference (RNAi)-based approaches, CHOP was found to be a key regulator of these responses. These effects are similar to those manifested by lewisite suggesting that PAO could be used as a prototype of arsenicals to define the molecular pathogenesis of chemical injury.


Subject(s)
Arsenicals/immunology , Edema/immunology , Erythema/immunology , Inflammation/immunology , Keratinocytes/metabolism , Transcription Factor CHOP/metabolism , Animals , Cell Line , Cells, Cultured , Edema/chemically induced , Endoplasmic Reticulum Chaperone BiP , Erythema/chemically induced , Eukaryotic Initiation Factor-2/metabolism , Heat-Shock Proteins/metabolism , Humans , Inflammation/chemically induced , Keratinocytes/pathology , Mice , Mice, Knockout , Oxidative Stress , Patched-1 Receptor/genetics , Phenylbutyrates/metabolism , RNA, Small Interfering/genetics , Transcription Factor CHOP/genetics , Unfolded Protein Response
14.
Ann N Y Acad Sci ; 1378(1): 143-157, 2016 08.
Article in English | MEDLINE | ID: mdl-27636894

ABSTRACT

Arsenicals are highly reactive inorganic and organic derivatives of arsenic. These chemicals are very toxic and produce both acute and chronic tissue damage. On the basis of these observations, and considering the low cost and simple methods of their bulk syntheses, these agents were thought to be appropriate for chemical warfare. Among these, the best-known agent that was synthesized and weaponized during World War I (WWI) is Lewisite. Exposure to Lewisite causes painful inflammatory and blistering responses in the skin, lung, and eye. These chemicals also manifest systemic tissue injury following their cutaneous exposure. Although largely discontinued after WWI, stockpiles are still known to exist in the former Soviet Union, Germany, Italy, the United States, and Asia. Thus, access by terrorists or accidental exposure could be highly dangerous for humans and the environment. This review summarizes studies that describe the biological, pathophysiological, toxicological, and environmental effects of exposure to arsenicals, with a major focus on cutaneous injury. Studies related to the development of novel molecular pathobiology-based antidotes against these agents are also described.


Subject(s)
Arsenic Poisoning/metabolism , Arsenicals/administration & dosage , Chemical Warfare Agents/poisoning , Environmental Exposure/adverse effects , Animals , Arsenic Poisoning/drug therapy , Arsenic Poisoning/epidemiology , Chemical Warfare/trends , Dimercaprol/therapeutic use , Humans , Oxidative Stress/drug effects , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
15.
Arch Biochem Biophys ; 609: 39-50, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27638049

ABSTRACT

Arsenic is a mitochondrial toxin, and its derivatives, such as arsenic trioxide (ATO), can trigger endoplasmic reticulum (ER) and the associated unfolded protein response (UPR). Here, we show that arsenic induction of the UPR triggers ATF4, which is involved in regulating this ER-mitochondrial crosstalk that is important for the molecular pathogenesis of arsenic toxicity. Employing ATF4+/+ and ATF4-/- MEFs, we show that ATO induces UPR and impairs mitochondrial integrity in ATF4+/+ MEF cells which is largely ablated upon loss of ATF4. Following ATO treatment, ATF4 activates NADPH oxidase by promoting assembly of the enzyme components Rac-1/P47phox/P67phox, which generates ROS/superoxides. Furthermore, ATF4 is required for triggering Ca++/calpain/caspase-12-mediated apoptosis following ATO treatment. The IP3R inhibitor attenuates Ca++/calpain-dependent apoptosis, as well as reduces m-ROS and MMP disruption, suggesting that ER-mitochondria crosstalk involves IP3R-regulated Ca++ signaling. Blockade of m-Ca++ entry by inhibiting m-VDAC reduces ATO-mediated UPR in ATF4+/+ cells. Additionally, ATO treatment leads to p53-regulated mitochondrial apoptosis, where p53 phosphorylation plays a key role. Together, these findings indicate that ATO-mediated apoptosis is regulated by both ER and mitochondria events that are facilitated by ATF4 and the UPR. Thus, we describe novel mechanisms by which ATO orchestrates cytotoxic responses involving interplay of ER and mitochondria.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Arsenicals/chemistry , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , NADPH Oxidases/metabolism , Oxides/chemistry , Activating Transcription Factor 4/genetics , Animals , Arsenic Trioxide , Calcium/chemistry , Cell Line , Cell Survival , Endoplasmic Reticulum Stress , Fibroblasts/metabolism , Homeostasis , Mice , Oxidation-Reduction , Phosphorylation , Protein Binding , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxides/metabolism
16.
Am J Pathol ; 186(10): 2637-49, 2016 10.
Article in English | MEDLINE | ID: mdl-27528504

ABSTRACT

Lewisite is a potent arsenic-based chemical warfare agent known to induce painful cutaneous inflammation and blistering. Only a few modestly effective antidotes have so far been described in the literature. However, the discovery of effective antidotes for lewisite was hampered by the paucity of the exact molecular mechanism underlying its cutaneous pathogenesis. We investigated the molecular mechanism underlying lewisite-induced cutaneous blistering and inflammation and describe its novel antidotes. On the basis of our initial screening, we used a highly sensitive murine model that recapitulates the known human pathogenesis of arsenicals-induced cutaneous inflammation and blistering. Topically administered lewisite induced potent acute inflammation and microvesication in the skin of Ptch1(+/-)/SKH-1 mice. Even at a very low dose, lewisite up-regulates unfolded protein response signaling, inflammatory response, and apoptosis. These cutaneous lesions were associated with production of reactive oxygen species and extensive apoptosis of the epidermal keratinocytes. We confirmed that activation of reactive oxygen species-dependent unfolded protein response signaling is the underlying molecular mechanism of skin damage. Similar alterations were noticed in lewisite-treated cultured human skin keratinocytes. We discovered that chemical chaperone 4-phenyl butyric acid and antioxidant N-acetylcysteine, which significantly attenuate lewisite-mediated skin injury, can serve as potent antidotes. These data reveal a novel molecular mechanism underlying the cutaneous pathogenesis of lewisite-induced lesions. We also identified novel potential therapeutic targets for lewisite-mediated cutaneous injury.


Subject(s)
Antidotes/pharmacology , Antioxidants/pharmacology , Blister/drug therapy , Chemical Warfare Agents/adverse effects , Molecular Chaperones/pharmacology , Patched-1 Receptor/genetics , Acetylcysteine/pharmacology , Animals , Arsenicals/adverse effects , Blister/chemically induced , Blister/pathology , Disease Models, Animal , Female , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/pathology , Keratinocytes/metabolism , Male , Mice , Mice, Hairless , Mice, Inbred C57BL , Patched-1 Receptor/metabolism , Phenylbutyrates/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
17.
Toxicol Appl Pharmacol ; 308: 46-58, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27461142

ABSTRACT

Chronic arsenic exposure to humans is considered immunosuppressive with augmented susceptibility to several infectious diseases. The exact molecular mechanisms, however, remain unknown. Earlier, we showed the involvement of unfolded protein response (UPR) signaling in arsenic-mediated impairment of macrophage functions. Here, we show that activating transcription factor 4 (ATF4), a UPR transcription factor, regulates arsenic trioxide (ATO)-mediated dysregulation of macrophage functions. In ATO-treated ATF4(+/+) wild-type mice, a significant down-regulation of CD11b expression was associated with the reduced phagocytic functions of peritoneal and lung macrophages. This severe immuno-toxicity phenotype was not observed in ATO-treated ATF4(+/-) heterozygous mice. To confirm these observations, we demonstrated in Raw 264.7 cells that ATF4 knock-down rescues ATO-mediated impairment of macrophage functions including cytokine production, bacterial engulfment and clearance of engulfed bacteria. Sustained activation of ATF4 by ATO in macrophages induces apoptosis, while diminution of ATF4 expression protects against ATO-induced apoptotic cell death. Raw 264.7 cells treated with ATO also manifest dysregulated Ca(++) homeostasis. ATO induces Ca(++)-dependent calpain-1 and caspase-12 expression which together regulated macrophage apoptosis. Additionally, apoptosis was also induced by mitochondria-regulated pathway. Restoring ATO-impaired Ca(++) homeostasis in ER/mitochondria by treatments with the inhibitors of inositol 1,4,5-trisphosphate receptor (IP3R) and voltage-dependent anion channel (VDAC) attenuate innate immune functions of macrophages. These studies identify a novel role for ATF4 in underlying pathogenesis of macrophage dysregulation and immuno-toxicity of arsenic.


Subject(s)
Activating Transcription Factor 4/metabolism , Immunity, Innate/drug effects , Macrophages, Alveolar/drug effects , Oxides/toxicity , Animals , Arsenic Trioxide , Arsenicals , Calcium/metabolism , Cell Line , Cytokines/biosynthesis , Homeostasis , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
Oncotarget ; 6(34): 36789-814, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26413810

ABSTRACT

Nevoid basal cell carcinoma syndrome (NBCCS) is a rare autosomal dominant disorder that is due, in large measure, to aberrant Shh signaling driven by mutations in the tumor suppressor gene Ptch1. Here, we describe the development of Ptch1+/-/ SKH-1 mice as a novel model of this disease. These animals manifest many features of NBCCS, including developmental anomalies and are remarkably sensitive to both ultraviolet (UVB) and ionizing radiation that drive the development of multiple BCCs. Just as in patients with NBCCS, Ptch1+/-/SKH-1 also spontaneously develops BCCs and other neoplasms such as rhabdomyomas/rhabdomyosarcomas. Administration of smoothened inhibitors (vismodegib/itraconazole/cyclopamine) or non-steroidal anti-inflammatory drug (sulindac/sulfasalazine) each result in partial resolution of BCCs in these animals. However, combined administration of these agents inhibits the growth of UVB-induced BCCs by >90%. Employing small molecule- and decoy-peptide-based approaches we further affirm that complete remission of BCCs could only be achieved by combined inhibition of p50-NFκB/Bcl3 and Shh signaling. We posit that Ptch1+/-/SKH-1 mice are a novel and relevant animal model for NBCCS. Understanding mechanisms that govern genetic predisposition to BCCs should facilitate our ability to identify and treat NBCCS gene carriers, including those at risk for sporadic BCCs while accelerating development of novel therapeutic modalities for these patients.


Subject(s)
Basal Cell Nevus Syndrome/metabolism , Hedgehog Proteins/metabolism , NF-kappa B p50 Subunit/metabolism , Proto-Oncogene Proteins/metabolism , Skin Neoplasms/metabolism , Transcription Factors/metabolism , Animals , B-Cell Lymphoma 3 Protein , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/pathology , Disease Models, Animal , Female , Hedgehog Proteins/genetics , Humans , Male , Mice , Mice, Hairless , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B p50 Subunit/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/genetics
19.
Arch Toxicol ; 89(2): 243-58, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24825450

ABSTRACT

Inhibition mechanism(s) of protein kinase B/Akt1 and its consequences on related cell signaling were investigated in human neuroblastoma SH-SY5Y cells exposed to 4-hydroxy-trans-2-nonenal (4-HNE), one of the most reactive aldehyde by-products of lipid peroxidation. In silico data indicate that 4-HNE interacts with kinase domain of Akt1 with the total docking score of 6.0577 and also forms H-bond to Glu234 residue similar to highly potent Akt1 inhibitor imidazopiperidine analog 8b, in which the protonated imidazole nitrogen involves in two hydrogen bonds between Glu234 and Asp292. The strong hydrogen bonding with Glu234 and hydrophobic interactions with several residues, namely Leu156, Gly157, Val164, Ala177, Tyr229, Ala230, Met281 and Thr291, at the vicinity which is normally occupied by the ribose of ATP, appear to be the main causes of Akt1 inhibition and lead to the significant conformational change on this region of protein. Results of mutational docking prove that Glu234 plays a major role in 4-HNE-mediated Akt1 inhibition. In silico data on Akt inhibition were further validated by observing the down-regulated levels of phosphorylated (Thr308/Ser493) Akt1 as well as the altered levels of the downstream targets of pAkt, namely downregulated levels of pGSK3ß (Ser9), ß-catenin, Bcl2 and upregulated levels of pro-apoptotic markers, namely Bad, Bax, P(53) and caspase-9/3. The cellular fate of such pAkt inhibition was evidenced by increased reactive oxygen species, degraded nuclei, transferase dUTP nick end labeling positive cells and upregulated levels of pJNK1/2. We identified that 4-HNE-mediated Akt1 inhibition was due to the competitive inhibition of ATP by 4-HNE at the kinase domain of ATP binding sites.


Subject(s)
Adenosine Triphosphate/metabolism , Aldehydes/pharmacology , Apoptosis/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Binding Sites , Cell Line, Tumor , Humans , Hydrogen Bonding , Molecular Docking Simulation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism
20.
Mol Neurobiol ; 51(2): 791-807, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24859382

ABSTRACT

Differentiating neuronal cells derived from human umbilical cord blood stem cells have been used as an in vitro tool for the assessment of developmental neurotoxicity of monocrotophos (MCP), an organophosphate pesticide. The differentiating cells were exposed to MCP during the different stages of maturation, viz., days 2, 4, and 8, and changes in the makers of cell proliferation, neuronal differentiation, neuronal injuries, and receptors were studied. We found significant upregulation in the different MAPKs, apoptosis, and neurogenesis markers and downregulation in the cell proliferation markers during neuronal differentiation. We further identified significant upregulation in the expression of different MAPKs and proteins involved in oxidative stress, apoptosis, and calpain pathways in the mid-differentiating cells exposed to MCP. The upregulated levels of these proteins seem to be the main cause of alteration during the differentiation process towards apoptosis as a fine-tune of pro-apoptotic and anti-apoptotic proteins are desirable for the process of differentiation without apoptosis. The decreased acetylcholinesterase activity, dopaminergic, and cholinergic receptors and increased acetylcholine levels in the differentiating neuronal cells indicate the vulnerability of these cells towards MCP-induced neurotoxicity. Our data confirms that differentiating neuronal cells derived from human umbilical cord stem cells could be used as a powerful tool to assess the developmental neurotoxicity in human beings.


Subject(s)
Cell Differentiation/drug effects , Fetal Blood/cytology , Fetal Blood/drug effects , Monocrotophos/toxicity , Neurons/drug effects , Stem Cells/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Fetal Blood/physiology , Humans , Insecticides/toxicity , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/pathology , Neurons/physiology , Stem Cells/pathology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...