1
|
Shi H, Ding Y, Sun P, Lv Z, Wang C, Ma H, Lu J, Yu B, Li W, Wang C. Chemical approaches targeting the hurdles of hepatocyte transplantation: mechanisms, applications, and advances. Front Cell Dev Biol 2024; 12:1480226. [PMID: 39544361 PMCID: PMC11560891 DOI: 10.3389/fcell.2024.1480226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Hepatocyte transplantation (HTx) has been a novel cell-based therapy for severe liver diseases, as the donor livers for orthotopic liver transplantation are of great shortage. However, HTx has been confronted with two main hurdles: limited high-quality hepatocyte sources and low cell engraftment and repopulation rate. To cope with, researchers have investigated on various strategies, including small molecule drugs with unique advantages. Small molecules are promising chemical tools to modulate cell fate and function for generating high quality hepatocyte sources. In addition, endothelial barrier, immune responses, and low proliferative efficiency of donor hepatocytes mainly contributes to low cell engraftment and repopulation rate. Interfering these biological processes with small molecules is beneficial for improving cell engraftment and repopulation. In this review, we will discuss the applications and advances of small molecules in modulating cell differentiation and reprogramming for hepatocyte resources and in improving cell engraftment and repopulation as well as its underlying mechanisms.
Collapse
Affiliation(s)
- Huanxiao Shi
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Yi Ding
- Experimental Teaching Center, Naval Medical University, Shanghai, China
| | - Pingxin Sun
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Zhuman Lv
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Chunyan Wang
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Haoxin Ma
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Junyu Lu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Bing Yu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Wenlin Li
- Department of Cell Biology, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, China
| | - Chao Wang
- Department of Cell Biology, Naval Medical University, Shanghai, China
| |
Collapse
|
2
|
Maadurshni GB, Nagarajan M, Mahalakshmi B, Sivasubramanian J, Hemamalini V, Manivannan J. 4-Methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP) exposure induces hepatotoxicity and nephrotoxicity - role of oxidative stress, mitochondrial dysfunction and pathways of cytotoxicity. Toxicol Res (Camb) 2024; 13:tfae173. [PMID: 39417036 PMCID: PMC11474237 DOI: 10.1093/toxres/tfae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/24/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Objective Bisphenol A (BPA) is a ubiquitous pollutant worldwide and 4-Methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP) is considered a major active metabolite of BPA with a wide range of potent toxicological properties. However, its adverse outcome pathway (AOP) on the hepatic and renal system has not yet been explored. Methods Hence, the current study evaluated its effect on cell survival, oxidative stress, and apoptosis. In addition, the influence of signalling pathways on cytotoxicity and ROS generating enzymes (NOX2 and XO) on oxidative stress was explored by siRNA knockdown experiments. Further, its molecular interaction with SOD, CAT, and HSA (molecular docking and dynamics) was evaluated and validated with spectroscopy (fluorescence and FTIR) based methods. Results The outcome indicates that MBP exposure dose dependently increased the cytotoxic response, oxidative stress, and apoptosis in both hepatocytes and kidney cells. Further, MAPK signalling pathways and oxidative stress influenced the overall cytotoxic response in both cells. In addition, the stimulatory (NOX2 and XO) and inhibitory (SOD and CAT) effects of MBP were observed, along with a robust interaction with HSA. Conclusions The overall observation illustrates that MBP exposure adversely impacts hepatic and renal cells through oxidative stress and relevant molecular pathways which may connect the missing links during risk assessment of BPA.
Collapse
Affiliation(s)
| | - Manikandan Nagarajan
- Environmental Health and Toxicology Laboratory, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore - 641046, Tamil Nadu, India
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65212, United States of America
| | - Balamurali Mahalakshmi
- Environmental Health and Toxicology Laboratory, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore - 641046, Tamil Nadu, India
| | | | - Vedagiri Hemamalini
- Department of Bioinformatics, Bharathiar University, Coimbatore - 641046, Tamil Nadu, India
| | - Jeganathan Manivannan
- Environmental Health and Toxicology Laboratory, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore - 641046, Tamil Nadu, India
| |
Collapse
|
3
|
Ates I, Stuart C, Rathbone T, Barzi M, He G, Major AM, Shankar V, Lyman RA, Angner SS, Mackay TFC, Srinivasan S, Farris AB, Bissig KD, Cottle RN. Ex vivo gene editing and cell therapy for hereditary tyrosinemia type 1. Hepatol Commun 2024; 8:e0424. [PMID: 38668730 DOI: 10.1097/hc9.0000000000000424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/13/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND We previously demonstrated the successful use of in vivo CRISPR gene editing to delete 4-hydroxyphenylpyruvate dioxygenase (HPD) to rescue mice deficient in fumarylacetoacetate hydrolase (FAH), a disorder known as hereditary tyrosinemia type 1 (HT1). The aim of this study was to develop an ex vivo gene-editing protocol and apply it as a cell therapy for HT1. METHODS We isolated hepatocytes from wild-type (C57BL/6J) and Fah-/- mice and then used an optimized electroporation protocol to deliver Hpd-targeting CRISPR-Cas9 ribonucleoproteins into hepatocytes. Next, hepatocytes were transiently incubated in cytokine recovery media formulated to block apoptosis, followed by splenic injection into recipient Fah-/- mice. RESULTS We observed robust engraftment and expansion of transplanted gene-edited hepatocytes from wild-type donors in the livers of recipient mice when transient incubation with our cytokine recovery media was used after electroporation and negligible engraftment without the media (mean: 46.8% and 0.83%, respectively; p=0.0025). Thus, the cytokine recovery medium was critical to our electroporation protocol. When hepatocytes from Fah-/- mice were used as donors for transplantation, we observed 35% and 28% engraftment for Hpd-Cas9 ribonucleoproteins and Cas9 mRNA, respectively. Tyrosine, phenylalanine, and biochemical markers of liver injury normalized in both Hpd-targeting Cas9 ribonucleoprotein and mRNA groups independent of induced inhibition of Hpd through nitisinone, indicating correction of disease indicators in Fah-/- mice. CONCLUSIONS The successful liver cell therapy for HT1 validates our protocol and, despite the known growth advantage of HT1, showcases ex vivo gene editing using electroporation in combination with liver cell therapy to cure a disease model. These advancements underscore the potential impacts of electroporation combined with transplantation as a cell therapy.
Collapse
Affiliation(s)
- Ilayda Ates
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Callie Stuart
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Tanner Rathbone
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Mercedes Barzi
- Department of Pediatrics, Division of Medical Genetics, Alice and Y.T. Chen Center for Genetics and Genomics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gordon He
- Department of Pediatrics, Division of Medical Genetics, Alice and Y.T. Chen Center for Genetics and Genomics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Angela M Major
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Vijay Shankar
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Rachel A Lyman
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Sidney S Angner
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Trudy F C Mackay
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Shanthi Srinivasan
- Department of Medicine, Digestive Diseases Division, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alton Brad Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Karl-Dimiter Bissig
- Department of Pediatrics, Division of Medical Genetics, Alice and Y.T. Chen Center for Genetics and Genomics, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biomedical Engineering (BME) at the Duke University Pratt School of Engineering, Durham, North Carolina, USA
- Duke Cancer Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Renee N Cottle
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
4
|
Nagarajan M, Maadurshni GB, Manivannan J. Bisphenol A (BPA) exposure aggravates hepatic oxidative stress and inflammatory response under hypertensive milieu - Impact of low dose on hepatocytes and influence of MAPK and ER stress pathways. Food Chem Toxicol 2024; 183:114197. [PMID: 38029875 DOI: 10.1016/j.fct.2023.114197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 10/27/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
Human exposure to the hazardous chemical, Bisphenol A (BPA), is almost ubiquitous. Due to the prevalence of hypertension (CVD risk factor) in the aged human population, it is necessary to explore its adverse effect in hypertensive subjects. The current study exposed the Nω-nitro-l-arginine methyl ester (L-NAME) induced hypertensive Wistar rats to human exposure relevant low dose of BPA (50 μg/kg) for 30 days period. The liver biochemical parameters, histopathology, immunohistochemistry, gene expression (RT-qPCR), trace elements (ICP-MS), primary rat hepatocytes cell culture and metabolomic (1H NMR) assessments were performed. Results illustrate that BPA exposure potentiates/aggravates hypertension induced tissue abnormalities (hepatic fibrosis), oxidative stress, ACE activity, malfunction of the antioxidant system, lipid abnormalities and inflammatory factor (TNF-α and IL-6) expression. Also, in cells, BPA increased ROS generation, mitochondrial dysfunction and lipid peroxidation without any impact on cytotoxicity and caspase 3 and 9 activation. Notably, BPA exposure modulate lipid metabolism (cholesterol and fatty acid) in primary hepatocytes. Finally, the influence of ERK1/2, p38MAPK, ER stress and oxidative stress during relatively high dose of BPA elicited cytotoxicity was observed. Therefore, a precise hazardous risk investigation of BPA exposure in hypertensive populations is highly recommended.
Collapse
Affiliation(s)
- Manikandan Nagarajan
- Environmental Health and Toxicology Laboratory, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Jeganathan Manivannan
- Environmental Health and Toxicology Laboratory, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
5
|
Karaarslan U, Çolak M, Topal S, Atakul G, Soydan E, Çağlar A, Ağın H. The association between N-acetylcysteine treatment and hepatic healing in patients with non-acetaminophen-induced liver injury in pediatric intensive care: A single-center retrospective study. Arch Pediatr 2022; 29:140-144. [PMID: 35039188 DOI: 10.1016/j.arcped.2021.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/31/2020] [Accepted: 11/28/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The aim of this study was to determine the association between the use of intravenous N-acetylcysteine (NAC) and hepatic healing in pediatric intensive care unit (PICU) patients with non-acetaminophen-induced hepatic injury, except for acute liver failure. METHODS The data of patients who received intravenous NAC as adjuvant therapy for transaminase levels more than sixfold normal values during their PICU stay between 2010 and 2014 were retrospectively collected from the medical records database. The patients who did not receive NAC with elevated transaminase levels during their PICU stay between 2014 and 2018 were also collected as the standard of care (SOC) cohort. RESULTS More than 50% of the liver injuries were secondary to acute hypoxia, hypotension, sepsis, and inflammation. The median number of elevated transaminase period (ETP) days of the NAC and SOC groups were 5 (IQR: 4) and 4 (IQR: 4), respectively (p = 0.17). There was no significant difference between the groups in terms of minimum and maximum laboratory values during ETP. There was no significant difference in terms of ETP and maximum ALT levels between the NAC and SOC groups in the hypoxia-hypotension subgroup. CONCLUSION This study did not show an association between indirect measures of hepatic healing and post-insult use of NAC in pediatric liver injury in the PICU setting.
Collapse
Affiliation(s)
- Utku Karaarslan
- Department of Pediatric Intensive Care, Dr. Behcet Uz Children's Hospital, Konak, Izmir 35210, Turkey.
| | - Mustafa Çolak
- Department of Pediatric Intensive Care, Dr. Behcet Uz Children's Hospital, Konak, Izmir 35210, Turkey
| | - Sevgi Topal
- Department of Pediatric Intensive Care, Dr. Behcet Uz Children's Hospital, Konak, Izmir 35210, Turkey
| | - Gülhan Atakul
- Department of Pediatric Intensive Care, Dr. Behcet Uz Children's Hospital, Konak, Izmir 35210, Turkey
| | - Ekin Soydan
- Department of Pediatric Intensive Care, Dr. Behcet Uz Children's Hospital, Konak, Izmir 35210, Turkey
| | - Aykut Çağlar
- Department of Pediatric Emergency Care, Dr. Behcet Uz Children's Hospital, Izmir, Turkey
| | - Hasan Ağın
- Department of Pediatric Intensive Care, Dr. Behcet Uz Children's Hospital, Konak, Izmir 35210, Turkey
| |
Collapse
|
6
|
Tang L, Wang F, Xiao L, Shen M, Xia S, Zhang Z, Zhang F, Zheng S, Tan S. Yi-Qi-Jian-Pi formula modulates the PI3K/AKT signaling pathway to attenuate acute-on-chronic liver failure by suppressing hypoxic injury and apoptosis in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114411. [PMID: 34265380 DOI: 10.1016/j.jep.2021.114411] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/01/2021] [Accepted: 07/11/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute-on-chronic liver failure (ACLF) is a key complication of chronic hepatitis, with a relatively high mortality rate and limited treatment options, which dramatically threatens human lives. Yi-Qi-Jian-Pi formula (YQJPF) is a herbal compound commonly used to treat liver failure. AIM OF THE STUDY The purpose of this research is to discuss the potential molecular biological effect and mechanism of YQJPF in ACLF. MATERIALS AND METHODS In this study, we created a rat model of ACLF by CCl4-, LPS- and D-Galactosamine (D-Gal) and an in vitro model of LPS-induced hepatocyte damage. The specific components of YQJPF and potential mechanism were explored based on bioinformatics analyses. Furthermore, we verified the effect of YQJPF on ACLF using immunohistochemistry, RT-qPCR, western blotting, and flow cytometry. RESULTS Our research demonstrated that, after YQJPF treatment, hepatocyte injury in rats was relieved. Bioinformatics analysis showed that PI3K/AKT, HIF-1, mitochondrial apoptosis pathways played prominent roles. YQJPF promoted HIF-1α protein expression and exerted protective effects against hypoxic injury, simultaneously reducing mitochondrial ROS production, suppressing hepatocyte apoptosis. Furthermore, we showed that YQJPF accelerates PI3K/AKT pathway activation, a known broad-spectrum inhibitor of PI3K. LY294002, which was used for reverse verification, suppressed the effect of YQJPF on hypoxic injury and ROS-mediated hepatocyte apoptosis. CONCLUSIONS YQJPF ameliorates liver injury by suppressing hypoxic injury and ROS-mediated hepatocyte apoptosis by modulating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Li Tang
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China; Department of Gastroenterology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China.
| | - Feixia Wang
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lingyan Xiao
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Min Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Siwei Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shanzhong Tan
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| |
Collapse
|
7
|
Zhang D, Cao Q, Jing L, Zhao X, Ma H. Establishment of a hypobaric hypoxia-induced cell injury model in PC12 cells. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:614-620. [PMID: 34986528 PMCID: PMC8732250 DOI: 10.3724/zdxbyxb-2021-0343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 08/10/2021] [Indexed: 11/25/2022]
Abstract
To construct a hypobaric hypoxia-induced cell injury model. Rat pheochromocytoma PC12 cells were randomly divided into control group, normobaric hypoxia group and hypobaric hypoxia group. The cells in control group were cultured at normal condition, while cells in other two groups were cultured in normobaric hypoxia and hypobaric hypoxia conditions, respectively. CCK-8 method was used to detect cell viability to determine the optimal modeling conditions like the oxygen concentration, atmospheric pressure and low-pressure hypoxia time. The contents of lactate dehydrogenase (LDH), superoxide dismutase (SOD) and malondialdehyde (MDA) were detected by microplate method. The apoptosis ratio and cell cycle were analyzed by flow cytometry. The hypobaric hypoxia-induced cell injury model can be established by culturing for 24 h at 1% oxygen concentration and 41 kPa atmospheric pressure. Compared with the control group and normobaric hypoxia group, the activity of LDH and the content of MDA in hypobaric hypoxia group were significantly increased, the activity of SOD was decreased, the percentage of apoptosis was increased (all <0.05), and the cell cycle was arrested in G0/G1 phase. A stable and reliable cell injury model induced by hypobaric hypoxia has been established with PC12 cells, which provides a suitable cell model for the experimental study on nerve injury induced by hypoxia at high altitude.
Collapse
Affiliation(s)
- Dongmei Zhang
- 2. Second Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention, Western War Zone, Chengdu 610000, China
| | - Qilu Cao
- 2. Second Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention, Western War Zone, Chengdu 610000, China
| | - Linlin Jing
- 2. Second Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention, Western War Zone, Chengdu 610000, China
| | - Xiuhua Zhao
- 2. Second Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention, Western War Zone, Chengdu 610000, China
| | - Huiping Ma
- 2. Second Department of Infectious Disease Control and Prevention, Center for Disease Control and Prevention, Western War Zone, Chengdu 610000, China
| |
Collapse
|
8
|
Shah J, Muir J, Furfaro D, Beitler JR, Dzierba AL. Use of N-Acetylcysteine for Clozapine-Induced Acute Liver Injury: A Case Report and Literature Review. J Pharm Pract 2021; 36:463-467. [PMID: 34284670 DOI: 10.1177/08971900211034007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Purpose: To report a case of clozapine-induced hepatotoxicity managed with intravenous (IV) N-acetylcysteine (NAC) and summarize the available literature. Summary: A 46-year-old woman with history of bipolar disorder with psychotic features presented to the intensive care unit with asterixis and elevations in liver enzymes. The patient had been initiated on risperidone, clozapine, and lithium approximately 1 month prior to admission. After ruling out other possible non-drug etiologies, clozapine was suspected as the likeliest cause of the acute liver injury. Her acute liver injury was managed with the discontinuation of all antipsychotics, administration of IV NAC, and other standard of care supportive measures. Conclusion: Although clozapine has been associated with hepatitis and acute liver failure, there are no reports of NAC used in the management of clozapine-induced hepatotoxicity. NAC was used in our patient after considering the potential benefit and limited adverse effects. The role of NAC in non-acetaminophen-induced acute liver failure remains promising, but more research is warranted.
Collapse
Affiliation(s)
- Jenny Shah
- Department of Pharmacy, 3740UPMC Pinnacle, Harrisburg, PA, USA
| | - Justin Muir
- Department of Pharmacy, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital, New York, NY, USA
| | - David Furfaro
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, NewYork-Presbyterian Hospital, New York, NY, USA
| | - Jeremy R Beitler
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, NewYork-Presbyterian Hospital, New York, NY, USA.,Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons, NewYork-Presbyterian Hospital, New York, NY, USA
| | - Amy L Dzierba
- Department of Pharmacy, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital, New York, NY, USA.,Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons, NewYork-Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
9
|
Ma M, Wu CJ, Zhang P, Li T, Wei SZ, Yu BT, Qin F, Yuan JH. N-acetylcysteine maintains penile length and erectile function in bilateral cavernous nerve crush rat model by reducing penile fibrosis. Asian J Androl 2021; 23:215-221. [PMID: 32394901 PMCID: PMC7991820 DOI: 10.4103/aja.aja_17_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Penile length shortening and erectile dysfunction are common complications after radical prostatectomy. Various methods have been used to maintain erectile function, but less attention has been paid to preserving penis length. N-acetylcysteine (NAC) has the effect of antioxidation and antifibrotic, which may be beneficial to improve those postoperative complications. This study investigated the effect of NAC on maintaining the penile length and the erectile function after bilateral cavernous nerve crush (BCNC) and its underlying mechanism. Twenty-four male rats were randomly divided into three groups: control group, BCNC group, and BCNC + NAC group. NAC or equal volume of saline was daily administrated by intragastric gavage for 4 weeks. The initial and end penile lengths were measured. Intracavernosal pressure/mean arterial pressure (ICP/MAP) ratio was calculated to assess erectile function. Hematoxylin–eosin staining, Masson's trichrome staining, immunohistochemistry, and Western blot were performed to explore cellular and molecular changes of the penis. Compared to the BCNC group, the penile length, ICP/MAP ratio and smooth muscle/collagen ratio in the BCNC + NAC group were improved significantly (all P < 0.05), and the expressions of endothelial nitric oxide synthase, α-smooth muscle actin, glutathione, and glutathione peroxidase 1 were significantly increased after NAC treated (all P < 0.05), along with the decreased expressions of hypoxia-inducible factor-1α, transforming growth factor-β1, collagen I, collagen III, collagen IV, malonaldehyde, and lysine oxidase (all P < 0.05). This study demonstrated that NAC could maintain penile length and partly improve erectile function. Possible mechanism is directly and/or indirectly related to antihypoxic and antifibrosis.
Collapse
Affiliation(s)
- Ming Ma
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chang-Jing Wu
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Zhang
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Li
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan-Zun Wei
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo-Tao Yu
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Qin
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiu-Hong Yuan
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|