1
|
Patel A, Rasheed A, Reilly I, Pareek Z, Hansen M, Haque Z, Simon-Fajardo D, Davies C, Tummala A, Reinhardt K, Bustabad A, Shaw M, Robins J, Vera Gomez K, Suphakorn T, Camacho Gemelgo M, Law A, Lin K, Hospedales E, Haley H, Perez Martinez JP, Khan S, DeCanio J, Padgett M, Abramov A, Nanjundan M. Modulation of Cytoskeleton, Protein Trafficking, and Signaling Pathways by Metabolites from Cucurbitaceae, Ericaceae, and Rosaceae Plant Families. Pharmaceuticals (Basel) 2022; 15:1380. [PMID: 36355554 PMCID: PMC9698530 DOI: 10.3390/ph15111380] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 10/22/2023] Open
Abstract
One promising frontier within the field of Medical Botany is the study of the bioactivity of plant metabolites on human health. Although plant metabolites are metabolic byproducts that commonly regulate ecological interactions and biochemical processes in plant species, such metabolites also elicit profound effects on the cellular processes of human and other mammalian cells. In this regard, due to their potential as therapeutic agents for a variety of human diseases and induction of toxic cellular responses, further research advances are direly needed to fully understand the molecular mechanisms induced by these agents. Herein, we focus our investigation on metabolites from the Cucurbitaceae, Ericaceae, and Rosaceae plant families, for which several plant species are found within the state of Florida in Hillsborough County. Specifically, we compare the molecular mechanisms by which metabolites and/or plant extracts from these plant families modulate the cytoskeleton, protein trafficking, and cell signaling to mediate functional outcomes, as well as a discussion of current gaps in knowledge. Our efforts to lay the molecular groundwork in this broad manner hold promise in supporting future research efforts in pharmacology and drug discovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Meera Nanjundan
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| |
Collapse
|
2
|
Li Y, Wang D, Ge H, Güngör C, Gong X, Chen Y. Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties. Pharmaceuticals (Basel) 2022; 15:1369. [PMID: 36355541 PMCID: PMC9698833 DOI: 10.3390/ph15111369] [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: 10/09/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 08/08/2023] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells possessing stemness characteristics that are closely associated with tumor proliferation, recurrence and resistance to therapy. Recent studies have shown that different cytoskeletal components and remodeling processes have a profound impact on the behavior of CSCs. In this review, we outline the different cytoskeletal components regulating the properties of CSCs and discuss current and ongoing therapeutic strategies targeting the cytoskeleton. Given the many challenges currently faced in targeted cancer therapy, a deeper comprehension of the molecular events involved in the interaction of the cytoskeleton and CSCs will help us identify more effective therapeutic strategies to eliminate CSCs and ultimately improve patient survival.
Collapse
Affiliation(s)
- Yuqiang Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Dan Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Heming Ge
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Cenap Güngör
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Xuejun Gong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yongheng Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
3
|
Chen F, Zhong Z, Zhang C, Lu Y, Chan YT, Wang N, Zhao D, Feng Y. Potential Focal Adhesion Kinase Inhibitors in Management of Cancer: Therapeutic Opportunities from Herbal Medicine. Int J Mol Sci 2022; 23:13334. [PMID: 36362132 PMCID: PMC9659249 DOI: 10.3390/ijms232113334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 08/15/2024] Open
Abstract
Focal adhesion kinase (FAK) is a multifunctional protein involved in cellular communication, integrating and transducing extracellular signals from cell-surface membrane receptors. It plays a central role intracellularly and extracellularly within the tumor microenvironment. Perturbations in FAK signaling promote tumor occurrence and development, and studies have revealed its biological behavior in tumor cell proliferation, migration, and adhesion. Herein we provide an overview of the complex biology of the FAK family members and their context-dependent nature. Next, with a focus on cancer, we highlight the activities of FAK signaling in different types of cancer and how knowledge of them is being used for screening natural compounds used in herbal medicine to fight tumor development.
Collapse
Affiliation(s)
- Feiyu Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Zhangfeng Zhong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yuanjun Lu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yau-Tuen Chan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Di Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Wurz AI, Schulz AM, O’Bryant CT, Sharp JF, Hughes RM. Cytoskeletal dysregulation and neurodegenerative disease: Formation, monitoring, and inhibition of cofilin-actin rods. Front Cell Neurosci 2022; 16:982074. [PMID: 36212686 PMCID: PMC9535683 DOI: 10.3389/fncel.2022.982074] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The presence of atypical cytoskeletal dynamics, structures, and associated morphologies is a common theme uniting numerous diseases and developmental disorders. In particular, cytoskeletal dysregulation is a common cellular feature of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. While the numerous activators and inhibitors of dysregulation present complexities for characterizing these elements as byproducts or initiators of the disease state, it is increasingly clear that a better understanding of these anomalies is critical for advancing the state of knowledge and plan of therapeutic attack. In this review, we focus on the hallmarks of cytoskeletal dysregulation that are associated with cofilin-linked actin regulation, with a particular emphasis on the formation, monitoring, and inhibition of cofilin-actin rods. We also review actin-associated proteins other than cofilin with links to cytoskeleton-associated neurodegenerative processes, recognizing that cofilin-actin rods comprise one strand of a vast web of interactions that occur as a result of cytoskeletal dysregulation. Our aim is to present a current perspective on cytoskeletal dysregulation, connecting recent developments in our understanding with emerging strategies for biosensing and biomimicry that will help shape future directions of the field.
Collapse
Affiliation(s)
- Anna I. Wurz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Anna M. Schulz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Collin T. O’Bryant
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Josephine F. Sharp
- Department of Chemistry, Notre Dame College, South Euclid, OH, United States
| | - Robert M. Hughes
- Department of Chemistry, East Carolina University, Greenville, NC, United States
- *Correspondence: Robert M. Hughes,
| |
Collapse
|
5
|
Kriek N, Nock SH, Sage T, Khalifa B, Bye AP, Mitchell JL, Thomson S, McLaughlin MG, Jones S, Gibbins JM, Unsworth AJ. Cucurbitacins Elicit Anti-Platelet Activity via Perturbation of the Cytoskeleton and Integrin Function. Thromb Haemost 2022; 122:1115-1129. [PMID: 35253142 PMCID: PMC9385249 DOI: 10.1055/a-1788-5322] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/06/2021] [Indexed: 11/23/2022]
Abstract
Cucurbitacins are dietary compounds that have been shown to elicit a range of anti-tumour, anti-inflammatory and anti-atherosclerotic activities. Originally identified as signal transducer and activator of transcription, STAT, inhibitors, a variety of mechanisms of action have since been described, including dysregulation of the actin cytoskeleton and disruption of integrin function. Integrin outside-in signalling and cytoskeletal rearrangements are critical for the propagation of stable thrombus formation and clot retraction following platelet adhesion at the site of vessel damage. The effects of cucurbitacins on platelet function and thrombus formation are unknown. We report for the first time anti-platelet and anti-thrombotic effects of cucurbitacins B, E and I in human platelets. Treatment of platelets with cucurbitacins resulted in attenuation of platelet aggregation, secretion and fibrinogen binding following stimulation by platelet agonists. Cucurbitacins were also found to potently inhibit other integrin- and cytoskeleton-mediated events, including adhesion, spreading and clot retraction. Further investigation of cytoskeletal dynamics found treatment with cucurbitacins altered cofilin phosphorylation, enhanced activation and increased F actin polymerisation and microtubule assembly. Disruption to cytoskeletal dynamics has been previously shown to impair integrin activation, platelet spreading and clot retraction. Anti-platelet properties of cucurbitacins were found to extend to a disruption of stable thrombus formation, with an increase in thrombi instability and de-aggregation under flow. Our research identifies novel, anti-platelet and anti-thrombotic actions of cucurbitacins that appear to be linked to dysregulation of cytoskeletal dynamics and integrin function.
Collapse
Affiliation(s)
- Neline Kriek
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Sophie H. Nock
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Tanya Sage
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Badrija Khalifa
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alexander P. Bye
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Joanne L. Mitchell
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Steven Thomson
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mark G. McLaughlin
- Department of Chemistry, Lancaster University, Lancaster, United Kingdom
| | - Sarah Jones
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Amanda J. Unsworth
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
6
|
Rather IIG, Behl T, Sehgal A, Singh S, Sharma N, Sharma A, Bhatia S, Al-Harrasi A, Khan N, Khan H, Bungau S. Exploration of potential role of Rho GTPase in nicotine dependence-induced withdrawal syndrome in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:17417-17424. [PMID: 34665416 DOI: 10.1007/s11356-021-17059-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
The RhoA gene showed an important genotypic association with nicotine dependence and smoking initiation. The current study aims to investigate the effect of the Rho GTPase inhibitor ML141 in the progression of nicotine dependence in a mice model of precipitated nicotine withdrawal syndrome by mecamylamine.The experimental procedure involved administration of 2.5 mg/kg nicotine dissolved in normal saline subcutaneously (s.c) four times a day consecutively for 7 days and last single dose in the morning on 8th day. ML-141 was dissolved in dimethyl sulfoxide (DMSO) and was administered daily with nicotine as corrective treatment at a dose of 1,5 and 10 mg/kg (p < 0.05). An injection of 3 mg/kg of mecamylamine intraperitoneal (ip) was given an hour later than the last nicotine dose on the day 8 to precipitate withdrawal of nicotine and withdrawal severity was assessed by measuring hyperalgesia, piloerection, jumping frequency, tremors, and withdrawal severity score (WSS). Various behavioural changes such as hyperalgesia, piloerection, jumping frequency, and tremors were monitored and WSS was calculated. ML-141 a selective Rho GTPase inhibitor was found to show dose-dependent effect on all these parameters. Inhibition of Rho GTPase was found to reduce the severity of withdrawal syndrome; therefore, it can be concluded that Rho GTPase would serve as a suitable biological target by regulating the reward system in brain and could be used as new target for drug discovery.
Collapse
Affiliation(s)
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Saurabh Bhatia
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | | | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
7
|
Chen Y, Peng S, Zhao P, Chen L, Liu GS, Ouyang D, Luo Y, Chen Z. Cell-modified plasmonic interface for the signal-amplified detection of Cucurbitacin E. BIOMEDICAL OPTICS EXPRESS 2022; 13:274-283. [PMID: 35154870 PMCID: PMC8803011 DOI: 10.1364/boe.445679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 05/24/2023]
Abstract
Cucurbitacin E (CuE) plays an important role in anticancer, antichemical carcinogenesis, and body immunity, etc., and the detection of its concentration is meaningful to pharmacological studies and clinical applications. However, the small molecular weight of CuE makes direct detection difficult through a surface plasmon resonance (SPR) sensor. In this work, we propose a cells-amplified signal strategy at the plasmonic interface, realizing the detection of CuE with ultra-low concentration. The seeded HeLa cells are modified onto the surface of the SPR sensor, and a small amount of CuE can lead to the remarkable morphology change of cells and the release of cell-related substances onto the plamonic interface, thus significantly amplifying the signal. Experimental results show that by using an unmodified SPR sensor with the bulk refractive index sensitivity of 2367.3 nm/RIU (RIU: refractive index unit), there no effective signal can be detected during the CuE concentration range of 0-100 nM; whereas, employing the proposed strategy, the signal for CuE detection can be significantly enhanced, resulting in a high detection sensitivity of 0.6196 nm/nM, corresponding to a limit of detection of 45.2 pM (25.2 pg/mL). The proposed cells-based signal amplifying strategy shows great potential applications in drug screening or bio-sensing to small molecules with low concentration.
Collapse
Affiliation(s)
- Yaofei Chen
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Jinan University, Guangzhou 510632, China
- Contributed equally
| | - Shuihua Peng
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Jinan University, Guangzhou 510632, China
- Contributed equally
| | - Peili Zhao
- Pathology department, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Lei Chen
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Jinan University, Guangzhou 510632, China
| | - Gui-Shi Liu
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Jinan University, Guangzhou 510632, China
| | - Dongyun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yunhan Luo
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Jinan University, Guangzhou 510632, China
| | - Zhe Chen
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Jinan University, Guangzhou 510632, China
| |
Collapse
|
8
|
Guiler W, Koehler A, Boykin C, Lu Q. Pharmacological Modulators of Small GTPases of Rho Family in Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:661612. [PMID: 34054432 PMCID: PMC8149604 DOI: 10.3389/fncel.2021.661612] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Classical Rho GTPases, including RhoA, Rac1, and Cdc42, are members of the Ras small GTPase superfamily and play essential roles in a variety of cellular functions. Rho GTPase signaling can be turned on and off by specific GEFs and GAPs, respectively. These features empower Rho GTPases and their upstream and downstream modulators as targets for scientific research and therapeutic intervention. Specifically, significant therapeutic potential exists for targeting Rho GTPases in neurodegenerative diseases due to their widespread cellular activity and alterations in neural tissues. This study will explore the roles of Rho GTPases in neurodegenerative diseases with focus on the applications of pharmacological modulators in recent discoveries. There have been exciting developments of small molecules, nonsteroidal anti-inflammatory drugs (NSAIDs), and natural products and toxins for each classical Rho GTPase category. A brief overview of each category followed by examples in their applications will be provided. The literature on their roles in various diseases [e.g., Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Frontotemporal dementia (FTD), and Multiple sclerosis (MS)] highlights the unique and broad implications targeting Rho GTPases for potential therapeutic intervention. Clearly, there is increasing knowledge of therapeutic promise from the discovery of pharmacological modulators of Rho GTPases for managing and treating these conditions. The progress is also accompanied by the recognition of complex Rho GTPase modulation where targeting its signaling can improve some aspects of pathogenesis while exacerbating others in the same disease model. Future directions should emphasize the importance of elucidating how different Rho GTPases work in concert and how they produce such widespread yet different cellular responses during neurodegenerative disease progression.
Collapse
Affiliation(s)
| | | | | | - Qun Lu
- Department of Anatomy and Cell Biology, The Harriet and John Wooten Laboratory for Alzheimer’s and Neurogenerative Diseases Research, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
9
|
Mesas C, Fuel M, Martínez R, Prados J, Melguizo C, Porres JM. In vitro evidence of the antitumor capacity of Solanaceae and Cucurbitaceae in colon cancer: A systematic review. Crit Rev Food Sci Nutr 2021; 62:6293-6314. [PMID: 33739207 DOI: 10.1080/10408398.2021.1900058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Colon cancer is the fourth leading cause of cancer deaths around the world. Despite advances in understanding its etiology and in diagnosis and treatment, new therapeutic strategies are still required. In this sense, the Solanaceae and Cucurbitaceae families have been widely used to treat various pathologies, including cancer, for their bioactive components. The objective of this systematic review was to analyze the antitumor activity of the bioactive components present in extracts from Solanaceae and Cucurbitaceae families using different in in vitro models of colon cancer. 241 publications have been identified (published from January 2008 to January 2020) from different electronic data base. 44 articles were included, 26 of which examined the Solanaceae family. The antitumor activity exhibited by this family was due to the withanolide-type steroid compounds they harbor. 18 articles were related to the Cucurbitaceae family. This family is characterized by their production of cucurbitacin-type triterpenoid compounds and their derivatives, which confer antitumor activity. In conclusion, the different genera belonging to both families are an important source of bioactive compounds with relevant activity against colon cancer. More experimental and in vivo studies will be required to corroborate their antitumor activity and to leverage them in future clinical practice.
Collapse
Affiliation(s)
- Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Marco Fuel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Rosario Martínez
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Jesús M Porres
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
10
|
Abstract
Simple Summary Cell migration is an essential process from embryogenesis to cell death. This is tightly regulated by numerous proteins that help in proper functioning of the cell. In diseases like cancer, this process is deregulated and helps in the dissemination of tumor cells from the primary site to secondary sites initiating the process of metastasis. For metastasis to be efficient, cytoskeletal components like actin, myosin, and intermediate filaments and their associated proteins should co-ordinate in an orderly fashion leading to the formation of many cellular protrusions-like lamellipodia and filopodia and invadopodia. Knowledge of this process is the key to control metastasis of cancer cells that leads to death in 90% of the patients. The focus of this review is giving an overall understanding of these process, concentrating on the changes in protein association and regulation and how the tumor cells use it to their advantage. Since the expression of cytoskeletal proteins can be directly related to the degree of malignancy, knowledge about these proteins will provide powerful tools to improve both cancer prognosis and treatment. Abstract Successful metastasis depends on cell invasion, migration, host immune escape, extravasation, and angiogenesis. The process of cell invasion and migration relies on the dynamic changes taking place in the cytoskeletal components; actin, tubulin and intermediate filaments. This is possible due to the plasticity of the cytoskeleton and coordinated action of all the three, is crucial for the process of metastasis from the primary site. Changes in cellular architecture by internal clues will affect the cell functions leading to the formation of different protrusions like lamellipodia, filopodia, and invadopodia that help in cell migration eventually leading to metastasis, which is life threatening than the formation of neoplasms. Understanding the signaling mechanisms involved, will give a better insight of the changes during metastasis, which will eventually help targeting proteins for treatment resulting in reduced mortality and longer survival.
Collapse
|
11
|
Sumanth MS, Jacob SP, Abhilasha KV, Manne BK, Basrur V, Lehoux S, Campbell RA, Yost CC, McIntyre TM, Cummings RD, Weyrich AS, Rondina MT, Marathe GK. Different glycoforms of alpha-1-acid glycoprotein contribute to its functional alterations in platelets and neutrophils. J Leukoc Biol 2020; 109:915-930. [PMID: 33070381 DOI: 10.1002/jlb.3a0720-422r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/18/2020] [Accepted: 09/30/2020] [Indexed: 12/23/2022] Open
Abstract
Alpha-1-acid glycoprotein (AGP-1) is a positive acute phase glycoprotein with uncertain functions. Serum AGP-1 (sAGP-1) is primarily derived from hepatocytes and circulates as 12-20 different glycoforms. We isolated a glycoform secreted from platelet-activating factor (PAF)-stimulated human neutrophils (nAGP-1). Its peptide sequence was identical to hepatocyte-derived sAGP-1, but nAGP-1 differed from sAGP-1 in its chromatographic behavior, electrophoretic mobility, and pattern of glycosylation. The function of these 2 glycoforms also differed. sAGP-1 activated neutrophil adhesion, migration, and neutrophil extracellular traps (NETosis) involving myeloperoxidase, peptidylarginine deiminase 4, and phosphorylation of ERK in a dose-dependent fashion, whereas nAGP-1 was ineffective as an agonist for these events. Furthermore, sAGP-1, but not nAGP-1, inhibited LPS-stimulated NETosis. Interestingly, nAGP-1 inhibited sAGP-1-stimulated neutrophil NETosis. The discordant effect of the differentially glycosylated AGP-1 glycoforms was also observed in platelets where neither of the AGP-1 glycoforms alone stimulated aggregation of washed human platelets, but sAGP-1, and not nAGP-1, inhibited aggregation induced by PAF or ADP, but not by thrombin. These functional effects of sAGP-1 correlated with intracellular cAMP accumulation and phosphorylation of the protein kinase A substrate vasodilator-stimulated phosphoprotein and reduction of Akt, ERK, and p38 phosphorylation. Thus, the sAGP-1 glycoform limits platelet reactivity, whereas nAGP-1 glycoform also limits proinflammatory actions of sAGP-1. These studies identify new functions for this acute phase glycoprotein and demonstrate that the glycosylation of AGP-1 controls its effects on 2 critical cells of acute inflammation.
Collapse
Affiliation(s)
- Mosale Seetharam Sumanth
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Shancy P Jacob
- Department of Pediatrics, Division of Allergy and Immunology, University of Utah, Salt Lake City, Utah, USA
| | | | - Bhanu Kanth Manne
- Molecular Medicine Program, and Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Venkatesha Basrur
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sylvain Lehoux
- Beth Israel Deaconess Medical Center, Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert A Campbell
- Molecular Medicine Program, and Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Christian C Yost
- Molecular Medicine Program, and Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, Utah, USA.,Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Thomas M McIntyre
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Richard D Cummings
- Beth Israel Deaconess Medical Center, Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew S Weyrich
- Molecular Medicine Program, and Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Matthew T Rondina
- Molecular Medicine Program, and Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, Utah, USA.,The Geriatric Research Education and Clinical Center, Salt Lake City, Utah, USA.,Department of Internal Medicine, George E. Wahlen VAMC, Salt Lake City, Utah, USA
| | - Gopal K Marathe
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India.,Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| |
Collapse
|
12
|
Risinger AL, Du L. Targeting and extending the eukaryotic druggable genome with natural products: cytoskeletal targets of natural products. Nat Prod Rep 2020; 37:634-652. [PMID: 31764930 PMCID: PMC7797185 DOI: 10.1039/c9np00053d] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Covering: 2014-2019We review recent progress on natural products that target cytoskeletal components, including microtubules, actin, intermediate filaments, and septins and highlight their demonstrated and potential utility in the treatment of human disease. The anticancer efficacy of microtubule targeted agents identified from plants, microbes, and marine organisms is well documented. We highlight new microtubule targeted agents currently in clinical evaluations for the treatment of drug resistant cancers and the accumulating evidence that the anticancer efficacy of these agents is not solely due to their antimitotic effects. Indeed, the effects of microtubule targeted agents on interphase microtubules are leading to their potential for more mechanistically guided use in cancers as well as neurological disease. The discussion of these agents as more targeted drugs also prompts a reevaluation of our thinking about natural products that target other components of the cytoskeleton. For instance, actin active natural products are largely considered chemical probes and non-selective toxins. However, studies utilizing these probes have uncovered aspects of actin biology that can be more specifically targeted to potentially treat cancer, neurological disorders, and infectious disease. Compounds that target intermediate filaments and septins are understudied, but their continued discovery and mechanistic evaluations have implications for numerous therapeutic indications.
Collapse
Affiliation(s)
- April L Risinger
- The University of Texas Health Science Center at San Antonio, Department of Pharmacology, 7703 Floyd Curl Drive, San Antonio, Texas 78229, USA.
| | | |
Collapse
|
13
|
Liang J, Zhang XL, Yuan JW, Zhang HR, Liu D, Hao J, Ji W, Wu XZ, Chen D. Cucurbitacin B inhibits the migration and invasion of breast cancer cells by altering the biomechanical properties of cells. Phytother Res 2018; 33:618-630. [PMID: 30548720 DOI: 10.1002/ptr.6250] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/31/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022]
Abstract
Changes in cellular biomechanical properties affect cell migration and invasion. The natural compound Cucurbitacin B (CuB) has potent anticancer activity; however, the mechanism underlying its inhibitory effect on breast cancer metastasis needs further study. Here, we showed that low-dose CuB inhibited adhesion and altered the viscoelasticity of breast cancer cells, thereby, reducing cell deformability. In vitro and in vivo experiments proved that CuB effectively inhibited the migration and invasion of breast cancer cells. Further studies have found that CuB downregulated the expression of F-actin/vimentin/FAK/vinculin in breast cancer cells, altering the distribution and reorganization of cytoskeletal proteins in the cells. CuB inhibited signaling by the Rho family GTPases RAC1/CDC42/RhoA downstream of integrin. These findings indicate that CuB has been proven to mediate the reorganization and distribution of cytoskeletal proteins of breast cancer cells through RAC1/CDC42/RhoA signaling, which improves the mechanical properties of cell adhesion and deformation and consequently inhibits cell migration and invasion.
Collapse
Affiliation(s)
- Jing Liang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiao-Lan Zhang
- Institute Of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jin-Wei Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hao-Ran Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dan Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jian Hao
- Tumor hematology, Tianjin 4th Center Hospital, Tianjin, China
| | - Wei Ji
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xiong-Zhi Wu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dan Chen
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
14
|
Tian Y, Xu L, He Y, Xu X, Li K, Ma Y, Gao Y, Wei D, Wei L. Knockdown of RAC1 and VASP gene expression inhibits breast cancer cell migration. Oncol Lett 2018; 16:2151-2160. [PMID: 30008913 PMCID: PMC6036495 DOI: 10.3892/ol.2018.8930] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
The ability of tumor cells to migrate is biologically fundamental for tumorigenesis, growth, metastasis and invasion. The present study examined the role of Ras-related C3 botulinum toxin substrate (RAC1) and vasodilator-stimulated phosphoprotein (VASP) in breast cancer cell migration. According to data in Kaplan, Oncomine and The Cancer Genome Atlas, increased expression levels of RAC1 and VASP in breast cancer are associated with decreased cancer cell differentiation, advanced pathological stage and more aggressive tumor subtypes, while increased VASP mRNA expression levels are positively correlated with a poor prognosis in patients with breast cancer. The short hairpin (sh)RNA technique was employed to knock down the expression of RAC1 or VASP. Stable interference with the expression of RAC1 or VASP using RAC1-shRNA or VASP-shRNA, respectively, was established in MCF-7 breast cancer cells. In RAC1-shRNA or VASP-shRNA cells, the protein expression levels of RAC1 or VASP were significantly downregulated compared with control cells. The proliferation and migration rates of the RAC1-shRNA or VASP-shRNA cells were significantly lower compared with control cells. It was observed that the protein expression levels of VASP also decreased in RAC1-shRNA cells compared with control cells. The results revealed that RAC1 and VASP may serve important roles in promoting the migration of MCF-7 breast cancer cells, and that VASP may among the downstream signaling molecules associated with RAC1.
Collapse
Affiliation(s)
- Yihao Tian
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China.,Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Liu Xu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yanqi He
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xiaolong Xu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Kai Li
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yanbin Ma
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yang Gao
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Defei Wei
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Lei Wei
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
15
|
Garg S, Kaul SC, Wadhwa R. Cucurbitacin B and cancer intervention: Chemistry, biology and mechanisms (Review). Int J Oncol 2017; 52:19-37. [PMID: 29138804 DOI: 10.3892/ijo.2017.4203] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/23/2017] [Indexed: 11/06/2022] Open
Abstract
Cancer is one of the most important healthcare matters, with the worst prognosis but the best possibilities for scientific development. It is likely to increase in the future and cause global havoc designating it as an epidemic. Cancer development requires urgent intervention. Past few decades have witnessed extensive research to challenge carcinogenesis. Treatment involving synthetic discipline is often associated with severe adverse effects, or even worsened prognosis. Accordingly, newer economic and patient friendly molecules are warranted. Many natural substances have proved their potential so far. Cucurbitacin B against cancer and other diseases has achieved towering popularity among the researchers around the world, as detailed in the below sections with summarized tables. In line with the fascinating role of cucurbitacin B against various types of cancers, through various molecular signaling pathways, it is justifiable to propose cucurbitacin B as a mainline chemotherapy before the onset and after the diagnosis of cancer.
Collapse
Affiliation(s)
- Sukant Garg
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| | - Sunil C Kaul
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| | - Renu Wadhwa
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| |
Collapse
|
16
|
Soman KV, Stafford SJ, Pazdrak K, Wu Z, Luo X, White WI, Wiktorowicz JE, Calhoun WJ, Kurosky A. Activation of Human Peripheral Blood Eosinophils by Cytokines in a Comparative Time-Course Proteomic/Phosphoproteomic Study. J Proteome Res 2017; 16:2663-2679. [PMID: 28679203 DOI: 10.1021/acs.jproteome.6b00367] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Activated eosinophils contribute to airway dysfunction and tissue remodeling in asthma and thus are considered to be important factors in asthma pathology. We report here comparative proteomic and phosphoproteomic changes upon activation of eosinophils using eight cytokines individually and in selected cytokine combinations in time-course reactions. Differential protein and phosphoprotein expressions were determined by mass spectrometry after 2-dimensional gel electrophoresis (2DGE) and by LC-MS/MS. We found that each cytokine-stimulation produced significantly different changes in the eosinophil proteome and phosphoproteome, with phosphoproteomic changes being more pronounced and having an earlier onset. Furthermore, we observed that IL-5, GM-CSF, and IL-3 showed the greatest change in protein expression and phosphorylation, and this expression differed markedly from those of the other five cytokines evaluated. Comprehensive univariate and multivariate statistical analyses were employed to evaluate the comparative results. We also monitored eosinophil activation using flow cytometry (FC) analysis of CD69. In agreement with our proteomic studies, FC indicated that IL-5, GM-CSF, and IL-3 were more effective than the other five cytokines studied in stimulating a cell surface CD69 increase indicative of eosinophil activation. Moreover, selected combinations of cytokines revealed proteomic patterns with many proteins in common with single cytokine expression patterns but also showed a greater effect of the two cytokines employed, indicating a more complex signaling pathway that was reflective of a more typical inflammatory pathology.
Collapse
Affiliation(s)
- Kizhake V Soman
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch , Galveston, Texas 77555, United States.,Sealy Center for Molecular Medicine, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | - Susan J Stafford
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | - Konrad Pazdrak
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch , Galveston, Texas 77555, United States.,Sealy Center for Molecular Medicine, University of Texas Medical Branch , Galveston, Texas 77555, United States.,Institute for Translational Sciences, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | - Zheng Wu
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | - Xuemei Luo
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | - Wendy I White
- MedImmune LLC , One MedImmune Way, Gaithersburg, Maryland 20878, United States
| | - John E Wiktorowicz
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch , Galveston, Texas 77555, United States.,Sealy Center for Molecular Medicine, University of Texas Medical Branch , Galveston, Texas 77555, United States.,Institute for Translational Sciences, University of Texas Medical Branch , Galveston, Texas 77555, United States.,Institute for Human Immunity & Infection, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | - William J Calhoun
- Department of Internal Medicine, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | - Alexander Kurosky
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch , Galveston, Texas 77555, United States.,Sealy Center for Molecular Medicine, University of Texas Medical Branch , Galveston, Texas 77555, United States
| |
Collapse
|
17
|
Protein kinase A activation by the anti-cancer drugs ABT-737 and thymoquinone is caspase-3-dependent and correlates with platelet inhibition and apoptosis. Cell Death Dis 2017; 8:e2898. [PMID: 28661475 PMCID: PMC5520940 DOI: 10.1038/cddis.2017.290] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 12/14/2022]
Abstract
Chemotherapy-induced thrombocytopenia is a common bleeding risk in cancer patients and limits chemotherapy dose and frequency. Recent data from mouse and human platelets revealed that activation of protein kinase A/G (PKA/PKG) not only inhibited thrombin/convulxin-induced platelet activation but also prevented the platelet pro-coagulant state. Here we investigated whether or not PKA/PKG activation could attenuate caspase-dependent apoptosis induced by the anti-cancer drugs ABT-737 (the precursor of navitoclax) and thymoquinone (TQ), thereby potentially limiting chemotherapy-induced thrombocytopenia. This is particularly relevant as activation of cyclic nucleotide signalling in combination chemotherapy is an emerging strategy in cancer treatment. However, PKA/PKG-activation, as monitored by phosphorylation of Vasodilator-stimulated phosphoprotein (VASP), did not block caspase-3-dependent platelet apoptosis induced by the compounds. In contrast, both substances induced PKA activation themselves and PKA activation correlated with platelet inhibition and apoptosis. Surprisingly, ABT-737- and TQ-induced VASP-phosphorylation was independent of cAMP levels and neither cyclases nor phosphatases were affected by the drugs. In contrast, however, ABT-737- and TQ-induced PKA activation was blocked by caspase-3 inhibitors. In conclusion, we show that ABT-737 and TQ activate PKA in a caspase-3-dependent manner, which correlates with platelet inhibition and apoptosis and therefore potentially contributes to the bleeding risk in chemotherapy patients.
Collapse
|
18
|
Wang X, Tanaka M, Peixoto HS, Wink M. Cucurbitacins: elucidation of their interactions with the cytoskeleton. PeerJ 2017; 5:e3357. [PMID: 28584704 PMCID: PMC5452965 DOI: 10.7717/peerj.3357] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/26/2017] [Indexed: 01/25/2023] Open
Abstract
Cucurbitacins, a class of toxic tetracyclic triterpenoids in Cucurbitaceae, modulate many molecular targets. Here we investigated the interactions of cucurbitacin B, E and I with cytoskeletal proteins such as microtubule and actin filaments. The effects of cucurbitacin B, E and I on microtubules and actin filaments were studied in living cells (Hela and U2OS) and in vitro using GFP markers, immunofluorescence staining and in vitro tubulin polymerization assay. Cucurbitacin B, E and I apparently affected microtubule structures in living cells and cucurbitacin E inhibited tubulin polymerization in vitro with IC50 value of 566.91 ± 113.5 µM. Cucurbitacin E did not affect the nucleation but inhibited the growth phase and steady state during microtubule assembly in vitro. In addition, cucurbitacin B, E and I all altered mitotic spindles and induced the cell cycle arrest at G2/M phase. Moreover, they all showed potent effects on actin cytoskeleton by affecting actin filaments through the depolymerization and aggregation. The interactions of cucubitacin B, E and I with microtubules and actin filaments present new insights into their modes of action.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Mine Tanaka
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Herbenya Silva Peixoto
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| |
Collapse
|
19
|
Colonne PM, Winchell CG, Graham JG, Onyilagha FI, MacDonald LJ, Doeppler HR, Storz P, Kurten RC, Beare PA, Heinzen RA, Voth DE. Vasodilator-Stimulated Phosphoprotein Activity Is Required for Coxiella burnetii Growth in Human Macrophages. PLoS Pathog 2016; 12:e1005915. [PMID: 27711191 PMCID: PMC5053435 DOI: 10.1371/journal.ppat.1005915] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 09/05/2016] [Indexed: 11/18/2022] Open
Abstract
Coxiella burnetii is an intracellular bacterial pathogen that causes human Q fever, an acute flu-like illness that can progress to chronic endocarditis and liver and bone infections. Humans are typically infected by aerosol-mediated transmission, and C. burnetii initially targets alveolar macrophages wherein the pathogen replicates in a phagolysosome-like niche known as the parasitophorous vacuole (PV). C. burnetii manipulates host cAMP-dependent protein kinase (PKA) signaling to promote PV formation, cell survival, and bacterial replication. In this study, we identified the actin regulatory protein vasodilator-stimulated phosphoprotein (VASP) as a PKA substrate that is increasingly phosphorylated at S157 and S239 during C. burnetii infection. Avirulent and virulent C. burnetii triggered increased levels of phosphorylated VASP in macrophage-like THP-1 cells and primary human alveolar macrophages, and this event required the Cα subunit of PKA. VASP phosphorylation also required bacterial protein synthesis and secretion of effector proteins via a type IV secretion system, indicating the pathogen actively triggers prolonged VASP phosphorylation. Optimal PV formation and intracellular bacterial replication required VASP activity, as siRNA-mediated depletion of VASP reduced PV size and bacterial growth. Interestingly, ectopic expression of a phospho-mimetic VASP (S239E) mutant protein prevented optimal PV formation, whereas VASP (S157E) mutant expression had no effect. VASP (S239E) expression also prevented trafficking of bead-containing phagosomes to the PV, indicating proper VASP activity is critical for heterotypic fusion events that control PV expansion in macrophages. Finally, expression of dominant negative VASP (S157A) in C. burnetii-infected cells impaired PV formation, confirming importance of the protein for proper infection. This study provides the first evidence of VASP manipulation by an intravacuolar bacterial pathogen via activation of PKA in human macrophages. Q fever, caused by the intracellular bacterial pathogen Coxiella burnetii, is an aerosol-transmitted infection that can develop into life-threatening chronic infections such as endocarditis. The pathogen preferentially grows within alveolar macrophages in a phagolysosome-like compartment termed the parasitophorous vacuole (PV). C. burnetii actively manipulates host cAMP-dependent protein kinase (PKA) signaling to promote PV formation and cell survival. Identification of bacterial effector proteins that manipulate PKA and downstream target proteins is critical to fully understand pathogen-mediated signaling circuits and develop new therapeutic strategies. Here, we found that PKA controls vasodilator-stimulated phosphoprotein (VASP) activity to promote PV formation and bacterial replication. VASP regulates actin-based motility used by a subset of intracellular bacteria for propulsion through the host cell cytosol and into bystander cells. However, C. burnetii does not use actin-based motility and replicates throughout its life cycle within a membrane bound vacuole. Thus, this study provides the first evidence of VASP manipulation by an intravacuolar bacterial pathogen. Characterization of VASP function in PV formation and identification of additional PKA substrates that promote infection will provide new insight into host-pathogen interactions during Q fever.
Collapse
Affiliation(s)
- Punsiri M. Colonne
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Caylin G. Winchell
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Joseph G. Graham
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Frances I. Onyilagha
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Laura J. MacDonald
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Heike R. Doeppler
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Richard C. Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Hospital Research Institute, Little Rock, Arkansas, United States of America
| | - Paul A. Beare
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Robert A. Heinzen
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Daniel E. Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
20
|
Monzen S, Chiba M, Hosokawa Y. Genetic network profiles associated with established resistance to ionizing radiation in acute promyelocytic leukemia cells and their extracellular vesicles. Oncol Rep 2016; 35:749-56. [PMID: 26718911 DOI: 10.3892/or.2015.4471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/23/2015] [Indexed: 11/06/2022] Open
Abstract
Radiation-resistant acute promyelocytic leukemia (APL) cells present challenges to treatment, and the acquisition of resistance to ionizing radiation (IR) is a matter of clinical concern. However, little information is available on the behavior of radio-resistant APL in terms of gene expression profiles and intercellular communication. In this study, cDNA microarray and RT-PCR were used to analyze the intracellular genetic network and extracellular vesicles (EVs), respectively, in the established radio-resistant HL60 (Res-HL60) cell line. Significant changes in the expression of 7,309 known mRNAs were observed in Res-HL60 relative to control. In addition, 7 mRNAs were determined as targets because significant changes in the expression were observed using Ingenuity analysis software, confirming the quantitative RT-PCR. However, EVs from Res-HL60 cells did not include these target molecules. These results suggest that radio-resistant APL is regulated by the expression and suppression of specific molecules, and these molecules are not transferred between cells by EVs.
Collapse
Affiliation(s)
- Satoru Monzen
- Department of Radiological Life Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Mitsuru Chiba
- Department of Biomedical Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Yoichiro Hosokawa
- Department of Radiological Life Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| |
Collapse
|
21
|
Wang L, Li C, Lin Q, Zhang X, Pan H, Xu L, Shi Z, Ouyang D, He X. Cucurbitacin E suppresses cytokine expression in human Jurkat T cells through down-regulating the NF-κB signaling. Acta Biochim Biophys Sin (Shanghai) 2015; 47:459-465. [PMID: 25921411 DOI: 10.1093/abbs/gmv030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 03/08/2015] [Indexed: 12/15/2022] Open
Abstract
Cucurbitacin E (CucE), a triterpenoid isolated from Cucurbitaceae plants, has been shown to possess an anti-inflammatory or immunosuppressive activity in vitro and in vivo, yet the underlying mechanism has been incompletely understood. The aim of the present study was to explore its effect on cytokine expression and the underlying mechanism in human Jurkat T cells as a cellular model. The results showed that CucE significantly inhibited the production of interleukin-2, tumor necrosis factor-α, and interferon-γ in culture medium of cells treated with phorbol 12,13-dibutyrate (PDB) plus ionomycin (Ion). Furthermore, the mRNA levels of these cytokines in activated Jurkat T cells were also decreased upon CucE treatment, suggesting a potential modulatory effect on the critical signaling pathways for cytokine expression, including nuclear factor-κB (NF-κB) or mitogen-activated protein kinases (MAPKs). In support of its effect on the NF-κB signaling pathway, CucE decreased the phosphorylation levels of inhibitor of κB (IκB) and NF-κB/p65 in PDB + Ion-stimulated cells. Further supporting this, the nuclear translocation of NF-κB/p65 was significantly suppressed in response to PDB plus Ion stimulation in the presence of CucE. The phosphorylation of p38MAPK, c-Jun N-terminal kinase (JNK), and Erk1/2, however, was not decreased or slightly increased at some time points by CucE treatment. Collectively, these data suggest that CucE may exhibit immunosuppressive effect by attenuating critical cytokine expression through down-regulating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lixian Wang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China Key Laboratory of Physical and Training Adapted Control System, Guangdong Provincial Institute of Sports Science, Guangzhou 510663, China
| | - Chenguang Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiuru Lin
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xiaoyu Zhang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hao Pan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Lihui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zijian Shi
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Dongyun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xianhui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
22
|
Zha QB, Zhang XY, Lin QR, Xu LH, Zhao GX, Pan H, Zhou D, Ouyang DY, Liu ZH, He XH. Cucurbitacin E Induces Autophagy via Downregulating mTORC1 Signaling and Upregulating AMPK Activity. PLoS One 2015; 10:e0124355. [PMID: 25970614 PMCID: PMC4430304 DOI: 10.1371/journal.pone.0124355] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/01/2015] [Indexed: 12/25/2022] Open
Abstract
Cucurbitacins, the natural triterpenoids possessing many biological activities, have been reported to suppress the mTORC1/p70S6K pathway and to induce autophagy. However, the correlation between such activities is largely unknown. In this study, we addressed this issue in human cancer cells in response to cucurbitacin E (CuE) treatment. Our results showed that CuE induced autophagy as evidenced by the formation of LC3-II and colocalization of punctate LC3 with the lysosomal marker LAMP2 in HeLa and MCF7 cells. However, CuE induced much lower levels of autophagy in ATG5-knocked down cells and failed to induce autophagy in DU145 cells lacking functional ATG5 expression, suggesting the dependence of CuE-induced autophagy on ATG5. Consistent with autophagy induction, mTORC1 activity (as reflected by p70S6K and ULK1S758 phosphorylation) was inhibited by CuE treatment. The suppression of mTORC1 activity was further confirmed by reduced recruitment of mTOR to the lysosome, which is the activation site of mTORC1. In contrast, CuE rapidly activated AMPK leading to increased phosphorylation of its substrates. AMPK activation contributed to CuE-induced suppression of mTORC1/p70S6K signaling and autophagy induction, since AMPK knockdown diminished these effects. Collectively, our data suggested that CuE induced autophagy in human cancer cells at least partly via downregulation of mTORC1 signaling and upregulation of AMPK activity.
Collapse
Affiliation(s)
- Qing-Bing Zha
- College of Life Science and Technology, Jinan University, Guangzhou, China
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-Yu Zhang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qiu-Ru Lin
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Li-Hui Xu
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Gao-Xiang Zhao
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Hao Pan
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Dan Zhou
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Dong-Yun Ouyang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ze-Huan Liu
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xian-Hui He
- College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
23
|
Identifying microRNA-mRNA regulatory network in gemcitabine-resistant cells derived from human pancreatic cancer cells. Tumour Biol 2015; 36:4525-34. [PMID: 25722110 DOI: 10.1007/s13277-015-3097-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/09/2015] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is unresectable in over 80 % of patients owing to difficulty in early diagnosis. Chemotherapy is the most frequently adopted therapy for advanced pancreatic cancer. The development of drug resistance to gemcitabine (GEM), which is always used in standard chemotherapy, often results in therapeutic failure. However, the molecular mechanisms underlying the gemcitabine resistance remain unclear. Therefore, we sought to explore the microRNA-mRNA network that is associated with the development of gemcitabine resistance and to identify molecular targets for overcoming the gemcitabine resistance. By exposing SW1990 pancreatic cancer cells to long-term gemcitabine with increasing concentrations, we established a gemcitabine-resistant cell line (SW1990/GEM) with a high IC50 (the concentration needed for 50 % growth inhibition, 847.23 μM). The mRNA and microRNA expression profiles of SW1990 cells and SW1990/GEM cells were determined using RNA-seq analysis. By comparing the results in control SW1990 cells, 507 upregulated genes and 550 downregulated genes in SW1990/GEM cells were identified as differentially expressed genes correlated with gemcitabine sensitivity. Gene ontology (GO) analysis showed that the differentially expressed genes were related to diverse biological processes. The upregulated genes were mainly associated with drug response and apoptosis, and the downregulated genes were correlated with cell cycle progression and RNA splicing. Concurrently, the differentially expressed microRNAs, which are the important player in drug resistance development, were also examined in SW1990/GEM cells, and 56 differential microRNAs were identified. Additionally, the expression profiles of selected genes and microRNAs were confirmed by using Q-PCR assays. Furthermore, combining the differentially expressed microRNAs and mRNAs as well as the predicted targets for these microRNAs, a core microRNA-mRNA regulatory network was constructed, which included hub microRNAs, such as hsa-miR-643, hsa-miR-4644, hsa-miR-4650-5p, hsa-miR-4455, hsa-miR-1261, and hsa-miR-3676. The predicted targets of these hub microRNAs in the microRNA-mRNA network were also observed in the identified differential genes. As a result, a differential gene and microRNA expression pattern was constructed in gemcitabine-resistant pancreatic cancer cells. Therefore, these data may be useful for the detection and treatment of drug resistance in pancreatic cancer patients, and the microRNA-mRNA network-based analysis is expected to be more effective and provides deep insights into the molecular mechanism of drug resistance.
Collapse
|