1
|
Cho H, Huh KM, Shim MS, Cho YY, Lee JY, Lee HS, Kwon YJ, Kang HC. Selective delivery of imaging probes and therapeutics to the endoplasmic reticulum or Golgi apparatus: Current strategies and beyond. Adv Drug Deliv Rev 2024; 212:115386. [PMID: 38971180 DOI: 10.1016/j.addr.2024.115386] [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: 05/01/2024] [Revised: 06/14/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
To maximize therapeutic effects and minimize unwanted effects, the interest in drug targeting to the endoplasmic reticulum (ER) or Golgi apparatus (GA) has been recently growing because two organelles are distributing hubs of cellular building/signaling components (e.g., proteins, lipids, Ca2+) to other organelles and the plasma membrane. Their structural or functional damages induce organelle stress (i.e., ER or GA stress), and their aggravation is strongly related to diseases (e.g., cancers, liver diseases, brain diseases). Many efforts have been developed to image (patho)physiological functions (e.g., oxidative stress, protein/lipid-related processing) and characteristics (e.g., pH, temperature, biothiols, reactive oxygen species) in the target organelles and to deliver drugs for organelle disruption using organelle-targeting moieties. Therefore, this review will overview the structure, (patho)physiological functions/characteristics, and related diseases of the organelles of interest. Future direction on ER or GA targeting will be discussed by understanding current strategies and investigations on targeting, imaging/sensing, and therapeutic systems.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Kang Moo Huh
- Departments of Polymer Science and Engineering & Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea.
| |
Collapse
|
2
|
Deng C, Zhao X, Chen Y, Ai K, Zhang Y, Gong T, Zeng C, Lei G. Engineered Platelet Microparticle-Membrane Camouflaged Nanoparticles for Targeting the Golgi Apparatus of Synovial Fibroblasts to Attenuate Rheumatoid Arthritis. ACS NANO 2022; 16:18430-18447. [PMID: 36342327 DOI: 10.1021/acsnano.2c06584] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Synovial fibroblasts in rheumatoid arthritis (RA) joints mediate synovial hyperplasia, progressive joint destruction, and the potential spread of disease between joints by producing multiple pathogenic proteins. Here, we deliver all-trans retinoic acid (ATRA) to selectively down-regulate these pathogenic factors, with a Golgi-targeting platelet microparticle-mimetic nanoplatform (termed Gol-PMMNP) which comprises a nanoparticle core and a platelet microparticle membrane coating labeled with a Golgi apparatus-targeting peptide. Gol-PMMNPs are shown to target synovial fibroblasts derived from RA patients via integrin α2β1-mediated endocytosis and accumulate in the Golgi apparatus by retrograde transport. ATRA-loaded Gol-PMMNPs (ATRA-Gol-PMMNPs) cause structural disruption of the Golgi apparatus, leading to an efficient reduction of pathogenic protein secretion in RA synovial fibroblasts. In rats with collagen-induced arthritis, Gol-PMMNPs display an arthritic joint-specific distribution, and ATRA-Gol-PMMNPs effectively reduce concentrations of pathogenic factors therein, including inflammatory cytokines, chemokines, and matrix-degrading enzymes within these joints. Additionally, ATRA-Gol-PMMNP treatment results in inflammatory remission and decreased bone erosion in both arthritic and proximal joints. Furthermore, ATRA-Gol-PMMNPs induce negligible toxicity to major organs. Taken together, ATRA-Gol-PMMNPs inhibit the progression of RA through reducing the production of multiple pathogenic mediators by synovial fibroblasts.
Collapse
Affiliation(s)
- Caifeng Deng
- Department of Orthopaedics and Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuan Zhao
- Department of Orthopaedics and Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuxiao Chen
- Department of Orthopaedics and Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Yuqing Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
- The Mongan Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Chao Zeng
- Department of Orthopaedics and Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guanghua Lei
- Department of Orthopaedics and Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
3
|
Chang HY, Yang WY. Golgi quality control and autophagy. IUBMB Life 2022; 74:361-370. [PMID: 35274438 DOI: 10.1002/iub.2611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 02/12/2022] [Indexed: 11/09/2022]
Abstract
Organelles can easily be disrupted by intracellular and extracellular factors. Studies on ER and mitochondria indicate that a wide range of responses are elicited upon organelle disruption. One response thought to be of particular importance is autophagy. Cells can target entire organelles into autophagosomes for removal. This wholesale nature makes autophagy a robust means for eliminating compromised organelles. Recently, it was demonstrated that the Golgi apparatus is a substrate of autophagy. On the other hand, various reports have shown that components traffic away from the Golgi for elimination in an autophagosome-independent manner when the Golgi apparatus is stressed. Future studies will reveal how these different pieces of machinery coordinate to drive Golgi degradation. Quantitative measurements will be needed to determine how much autophagy contributes to the maintenance of the Golgi apparatus.
Collapse
Affiliation(s)
- Hsiang-Yi Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wei Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, College of Life Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
4
|
Chiu SC, Huang YRJ, Wei TYW, Chen JMM, Kuo YC, Huang YTJ, Liao YTA, Yu CTR. The PRMT5/HURP axis retards Golgi repositioning by stabilizing acetyl-tubulin and Golgi apparatus during cell migration. J Cell Physiol 2021; 237:1033-1043. [PMID: 34541678 DOI: 10.1002/jcp.30589] [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: 06/02/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 11/10/2022]
Abstract
The Golgi apparatus (GA) translocates to the cell leading end during directional migration, thereby determining cell polarity and transporting essential factors to the migration apparatus. The study provides mechanistic insights into how GA repositioning (GR) is regulated. We show that the methyltransferase PRMT5 methylates the microtubule regulator HURP at R122. The HURP methylation mimicking mutant 122F impairs GR and cell migration. Mechanistic studies revealed that HURP 122F or endogenous methylated HURP, that is, HURP m122, interacts with acetyl-tubulin. Overexpression of HURP 122F stabilizes the bundling pattern of acetyl-tubulin by decreasing the sensitivity of the latter to a microtubule disrupting agent nocodazole. HURP 122F also rigidifies GA via desensitizing the organelle to several GA disrupting chemicals. Similarly, the acetyl-tubulin mimicking mutant 40Q or tubulin acetyltransferase αTAT1 can rigidify GA, impair GR, and retard cell migration. Reversal of HURP 122F-induced GA rigidification, by knocking down GA assembly factors such as GRASP65 or GM130, attenuates 122F-triggered GR and cell migration. Remarkably, PRMT5 is found downregulated and the level of HURP m122 is decreased during the early hours of wound healing-based cell migration, collectively implying that the PRMT5-HURP-acetyl-tubulin axis plays the role of brake, preventing GR and cell migration before cells reach empty space.
Collapse
Affiliation(s)
- Shao-Chih Chiu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Medical Research, Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | | | - Tong-You Wade Wei
- Graduate Institute of Biomedicine and Biomedical Technology, National Chi Nan University, Nantou, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Jo-Mei Maureen Chen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Yi-Chun Kuo
- Graduate Institute of Biomedicine and Biomedical Technology, National Chi Nan University, Nantou, Taiwan
| | - Yu-Ting Jenny Huang
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Yu-Ting Amber Liao
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chang-Tze Ricky Yu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.,Graduate Institute of Biomedicine and Biomedical Technology, National Chi Nan University, Nantou, Taiwan
| |
Collapse
|
5
|
Directed Differentiation of Human Pluripotent Stem Cells towards Corneal Endothelial-Like Cells under Defined Conditions. Cells 2021; 10:cells10020331. [PMID: 33562615 PMCID: PMC7915025 DOI: 10.3390/cells10020331] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
The most crucial function of corneal endothelial cells (CEnCs) is to maintain optical transparency by transporting excess fluid out of stroma. Unfortunately, CEnCs are not able to proliferate in vivo in the case of trauma or dystrophy. Visually impaired patients with corneal endothelial deficiencies that are waiting for transplantation due to massive global shortage of cadaveric corneal transplants are in a great need of help. In this study, our goal was to develop a defined, clinically applicable protocol for direct differentiation of CEnCs from human pluripotent stem cells (hPSCs). To produce feeder-free hPSC-CEnCs, we used small molecule induction with transforming growth factor (TGF) beta receptor inhibitor SB431542, GSK-3-specific inhibitor CHIR99021 and retinoic acid to guide differentiation through the neural crest and periocular mesenchyme (POM). Cells were characterized by the morphology and expression of human (h)CEnC markers with immunocytochemistry and RT-qPCR. After one week of induction, we observed the upregulation of POM markers paired-like homeodomain transcription factor 2 (PITX2) and Forkhead box C1 (FOXC1) and polygonal-shaped cells expressing CEnC-associated markers Zona Occludens-1 (ZO-1), sodium-potassium (Na+/K+)-ATPase, CD166, sodium bicarbonate cotransporter 1 (SLC4A4), aquaporin 1 (AQP1) and N-cadherin (NCAD). Furthermore, we showed that retinoic acid induced a dome formation in the cell culture, with a possible indication of fluid transport by the differentiated cells. Thus, we successfully generated CEnC-like cells from hPSCs with a defined, simple and fast differentiation method.
Collapse
|
6
|
Guieu B, Jourdan JP, Dreneau A, Willand N, Rochais C, Dallemagne P. Desirable drug-drug interactions or when a matter of concern becomes a renewed therapeutic strategy. Drug Discov Today 2020; 26:315-328. [PMID: 33253919 DOI: 10.1016/j.drudis.2020.11.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/14/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
Abstract
Drug-drug interactions are sometimes considered to be detrimental and responsible for adverse effects. In some cases, however, some are stakeholders of the efficiency of the treatment and this combinatorial strategy is exploited by some drug associations, including levodopa (L-Dopa) and dopadecarboxylase inhibitors, β-lactam antibiotics and clavulanic acid, 5-fluorouracil (5-FU) and folinic acid, and penicillin and probenecid. More recently, some drug-drug combinations have been integrated in modern drug design strategies, aiming to enhance the efficiency of already marketed drugs with new compounds acting not only as synergistic associations, but also as real boosters of activity. In this review, we provide an update of examples of such strategies, with a special focus on microbiology and oncology.
Collapse
Affiliation(s)
- Benjamin Guieu
- Normandie University, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Jean-Pierre Jourdan
- Normandie University, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France; Department of Pharmacy, Caen University Hospital, Caen, F-14000, France
| | - Aurore Dreneau
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Nicolas Willand
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Christophe Rochais
- Normandie University, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Patrick Dallemagne
- Normandie University, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France.
| |
Collapse
|
7
|
Cao XW, Wang FJ, Liew OW, Lu YZ, Zhao J. Analysis of Triterpenoid Saponins Reveals Insights into Structural Features Associated with Potent Protein Drug Enhancement Effects. Mol Pharm 2020; 17:683-694. [PMID: 31913047 DOI: 10.1021/acs.molpharmaceut.9b01158] [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: 01/01/2023]
Abstract
Plant-based saponins are amphipathic glycosides composed of a hydrophobic aglycone backbone covalently bound to one or more hydrophilic sugar moieties. Recently, the endosomal escape activity of triterpenoid saponins has been investigated as a potentially powerful tool for improved cytosolic penetration of protein drugs internalized by endocytic uptake, thereby greatly enhancing their pharmacological effects. However, only a few saponins have been studied, and the paucity in understanding the structure-activity relationship of saponins imposes significant limitations on their applications. To address this knowledge gap, 12 triterpenoid saponins with diverse structural side chains were screened for their utility as endosomolytic agents. These compounds were used in combination with a toxin (MAP30-HBP) comprising a type I ribosome-inactivating protein fused to a cell-penetrating peptide. Suitability of saponins as endosomolytic agents was assessed on the basis of cytotoxicity, endosomal escape promotion, and synergistic effects on toxins. Five saponins showed strong endosomal escape activity, enhancing MAP30-HBP cytotoxicity by more than 106 to 109 folds. These saponins also enhanced the apoptotic effect of MAP30-HBP in a pH-dependent manner. Additionally, growth inhibition of MAP30-HBP-treated SMMC-7721 cells was greater than that of similarly treated HeLa cells, suggesting that saponin-mediated endosomolytic effect is likely to be cell-specific. Furthermore, the structural features and hydrophobicity of the sugar side chains were analyzed to draw correlations with endosomal escape activity and derive predictive rules, thus providing new insights into structure-activity relationships of saponins. This study revealed new saponins that can potentially be exploited as efficient cytosolic delivery reagents for improved therapeutic drug effects.
Collapse
Affiliation(s)
- Xue-Wei Cao
- Department of Applied Biology , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China
| | - Fu-Jun Wang
- New Drug R&D Center , Zhejiang Fonow Medicine Company, Ltd. , 209 West Hulian Road , Dongyang 322100 , Zhejiang , China.,Shanghai R&D Center for Standardization of Chinese Medicines , 1200 Cailun Road , Shanghai 201203 , China.,Institute of Chinese Materia , Shanghai University of Traditional Chinese Medicine , 1200 Cailun Road , Shanghai 201203 , China
| | - Oi-Wah Liew
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System , Centre for Translational Medicine , MD6#08-01, 14 Medical Drive , 117599 , Singapore
| | - Ye-Zhou Lu
- Department of Applied Biology , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China
| | - Jian Zhao
- Department of Applied Biology , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China.,State Key Laboratory of Bioreactor Engineering , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China
| |
Collapse
|
8
|
Li H, Zhang P, Luo J, Hu D, Huang Y, Zhang ZR, Fu Y, Gong T. Chondroitin Sulfate-Linked Prodrug Nanoparticles Target the Golgi Apparatus for Cancer Metastasis Treatment. ACS NANO 2019; 13:9386-9396. [PMID: 31375027 DOI: 10.1021/acsnano.9b04166] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Metastasis is a multistep biological process regulated by multiple signaling pathways. The integrity of the Golgi apparatus plays an important role in these signaling pathways. Inspired by the mechanism and our previous finding about accumulation of chondroitin sulfate in Golgi apparatus in hepatic stellate cells, we developed a Golgi apparatus-targeting prodrug nanoparticle system by synthesizing retinoic acid (RA)-conjugated chondroitin sulfate (CS) (CS-RA). The prodrug nanoparticles appeared to accumulate in the Golgi apparatus in cancer cells and realized RA release under an acidic environment. We confirmed that CS-RA exhibited successful inhibition of multiple metastasis-associated proteins expression in vitro and in vivo by disruption of the Golgi apparatus structure. Following loading with paclitaxel (PTX), the CS-RA based nanoformulation (PTX-CS-RA) inhibited migration, invasion, and angiogenesis in vitro and suppressed tumor growth and metastasis in 4T1-Luc bearing mice. This multistep targeted nanoparticle system potentially enhanced the effect of antimetastasis combined with chemotherapy.
Collapse
Affiliation(s)
- Haohuan Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Pei Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Jingwen Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Danrong Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Zhi-Rong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| |
Collapse
|
9
|
Disassembling a cancer puzzle: Cell junctions and plasma membrane as targets for anticancer therapy. J Control Release 2018; 286:125-136. [PMID: 30030181 DOI: 10.1016/j.jconrel.2018.07.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023]
Abstract
Despite an enhanced permeability and retention effect typical of many solid tumors, drug penetration is not always sufficient. Possible strategies for the drug delivery improvement are a modification of the tumor cell-to-cell junctions and usage of cell membrane permeabilization proteins. In this review we discuss epithelial cell junctions as targets for a combined anticancer therapy and propose new possible sources of such agents. We suggest considering viral and bacterial pathogens disrupting epithelial layers as plentiful sources of new therapeutic agents for increasing tumor permeability for other effector agents. We also observe the application of pore forming proteins and peptides of different origin for cytoplasmic delivery of anti-cancer agents and consider the main obstacles of their use in vivo.
Collapse
|
10
|
Augmenting the Efficacy of Immunotoxins and Other Targeted Protein Toxins by Endosomal Escape Enhancers. Toxins (Basel) 2016; 8:toxins8070200. [PMID: 27376327 PMCID: PMC4963833 DOI: 10.3390/toxins8070200] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/14/2016] [Accepted: 06/17/2016] [Indexed: 12/18/2022] Open
Abstract
The toxic moiety of almost all protein-based targeted toxins must enter the cytosol of the target cell to mediate its fatal effect. Although more than 500 targeted toxins have been investigated in the past decades, no antibody-targeted protein toxin has been approved for tumor therapeutic applications by the authorities to date. Missing efficacy can be attributed in many cases to insufficient endosomal escape and therefore subsequent lysosomal degradation of the endocytosed toxins. To overcome this drawback, many strategies have been described to weaken the membrane integrity of endosomes. This comprises the use of lysosomotropic amines, carboxylic ionophores, calcium channel antagonists, various cell-penetrating peptides of viral, bacterial, plant, animal, human and synthetic origin, other organic molecules and light-induced techniques. Although the efficacy of the targeted toxins was typically augmented in cell culture hundred or thousand fold, in exceptional cases more than million fold, the combination of several substances harbors new problems including additional side effects, loss of target specificity, difficulties to determine the therapeutic window and cell type-dependent variations. This review critically scrutinizes the chances and challenges of endosomal escape enhancers and their potential role in future developments.
Collapse
|
11
|
Human Cytolytic Fusion Proteins: Modified Versions of Human Granzyme B and Angiogenin Have the Potential to Replace Bacterial Toxins in Targeted Therapies against CD64+ Diseases. Antibodies (Basel) 2014. [DOI: 10.3390/antib3010092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
12
|
Gilabert-Oriol R, Weng A, Mallinckrodt BV, Melzig MF, Fuchs H, Thakur M. Immunotoxins constructed with ribosome-inactivating proteins and their enhancers: a lethal cocktail with tumor specific efficacy. Curr Pharm Des 2014; 20:6584-643. [PMID: 25341935 PMCID: PMC4296666 DOI: 10.2174/1381612820666140826153913] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 08/05/2014] [Indexed: 11/30/2022]
Abstract
The term ribosome-inactivating protein (RIP) is used to denominate proteins mostly of plant origin, which have N-glycosidase enzymatic activity leading to a complete destruction of the ribosomal function. The discovery of the RIPs was almost a century ago, but their usage has seen transition only in the last four decades. With the advent of antibody therapy, the RIPs have been a subject of extensive research especially in targeted tumor therapies, which is the primary focus of this review. In the present work we enumerate 250 RIPs, which have been identified so far. An attempt has been made to identify all the RIPs that have been used for the construction of immunotoxins, which are conjugates or fusion proteins of an antibody or ligand with a toxin. The data from 1960 onwards is reviewed in this paper and an extensive list of more than 450 immunotoxins is reported. The clinical reach of tumor-targeted toxins has been identified and detailed in the work as well. While there is a lot of potential that RIPs embrace for targeted tumor therapies, the success in preclinical and clinical evaluations has been limited mainly because of their inability to escape the endo/lysosomal degradation. Various strategies that can increase the efficacy and lower the required dose for targeted toxins have been compiled in this article. It is plausible that with the advancements in platform technologies or improved endosomal escape the usage of tumor targeted RIPs would see the daylight of clinical success.
Collapse
Affiliation(s)
| | | | | | | | | | - Mayank Thakur
- Institut fur Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charite - Universitatsmedizin Berlin, Campus Virchow-Klinikum (Forum 4), Augustenburger Platz 1, D-13353 Berlin, Germany.
| |
Collapse
|
13
|
Antignani A, FitzGerald D. Immunotoxins: the role of the toxin. Toxins (Basel) 2013; 5:1486-502. [PMID: 23965432 PMCID: PMC3760048 DOI: 10.3390/toxins5081486] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 07/30/2013] [Accepted: 08/06/2013] [Indexed: 02/07/2023] Open
Abstract
Immunotoxins are antibody-toxin bifunctional molecules that rely on intracellular toxin action to kill target cells. Target specificity is determined via the binding attributes of the chosen antibody. Mostly, but not exclusively, immunotoxins are purpose-built to kill cancer cells as part of novel treatment approaches. Other applications for immunotoxins include immune regulation and the treatment of viral or parasitic diseases. Here we discuss the utility of protein toxins, of both bacterial and plant origin, joined to antibodies for targeting cancer cells. Finally, while clinical goals are focused on the development of novel cancer treatments, much has been learned about toxin action and intracellular pathways. Thus toxins are considered both medicines for treating human disease and probes of cellular function.
Collapse
Affiliation(s)
- Antonella Antignani
- Authors to whom correspondence should be addressed; E-Mail: (A.A.); (D.F.); Tel.: +1-301-496-9457 (D.F.); Fax: +1-301-402-1344 (D.F.)
| | - David FitzGerald
- Authors to whom correspondence should be addressed; E-Mail: (A.A.); (D.F.); Tel.: +1-301-496-9457 (D.F.); Fax: +1-301-402-1344 (D.F.)
| |
Collapse
|
14
|
Pirie CM, Liu DV, Wittrup KD. Targeted cytolysins synergistically potentiate cytoplasmic delivery of gelonin immunotoxin. Mol Cancer Ther 2013; 12:1774-82. [PMID: 23832121 DOI: 10.1158/1535-7163.mct-12-1023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Targeted endocytic uptake is a first step toward tissue-specific cytoplasmic macromolecular delivery; however, inefficient escape from the endolysosomal compartment makes this generally impractical at present. We report here a targeted cytolysin approach that dramatically potentiates endosomal release of an independently targeted potent gelonin immunotoxin. Fibronectin domains engineered for affinity to EGF receptor or carcinoembryonic antigen were fused to the plant toxin gelonin or bacterial pore-forming cytolysins. These fusion proteins display synergistic activity in both antigen-specific cytotoxicity in vitro, enhancing potency by several orders of magnitude, and in tumor growth inhibition in vivo. In addition, the number of internalized gelonin molecules required to induce apoptosis is reduced from approximately 5 × 10(6) to less than 10(3). Targeted potentiation shows promise for enhancing cytoplasmic delivery of other macromolecular payloads such as DNA, siRNA, and miRNA.
Collapse
Affiliation(s)
- Christopher M Pirie
- Corresponding Author: K. Dane Wittrup, Massachusetts Institute of Technology, Building 76-261, 77 Massachusetts Avenue, Cambridge, MA 02139.
| | | | | |
Collapse
|
15
|
Ricin and Ricin-Containing Immunotoxins: Insights into Intracellular Transport and Mechanism of action in Vitro. Antibodies (Basel) 2013. [DOI: 10.3390/antib2020236] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
16
|
Dissecting the Entry Route of Saporin-based a-CD7 Immunotoxins in Human T-Cell Acute Lymphoblastic Leukaemia Cells. Antibodies (Basel) 2013. [DOI: 10.3390/antib2010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
17
|
O'Hara JM, Yermakova A, Mantis NJ. Immunity to ricin: fundamental insights into toxin-antibody interactions. Curr Top Microbiol Immunol 2012; 357:209-41. [PMID: 22113742 PMCID: PMC4433546 DOI: 10.1007/82_2011_193] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ricin toxin is an extraordinarily potent inducer of cell death and inflammation. Ricin is also a potent provocateur of the humoral immune system, eliciting a mixture of neutralizing, non-neutralizing and even toxin-enhancing antibodies. The characterization of dozens of monoclonal antibodies (mAbs) against the toxin's enzymatic (RTA) and binding (RTB) subunits has begun to reveal fundamental insights into the underlying mechanisms by which antibodies neutralize (or fail to neutralize) ricin in systemic and mucosal compartments. This information has had immediate applications in the design, development and evaluation of ricin subunit vaccines and immunotherapeutics.
Collapse
Affiliation(s)
- Joanne M. O'Hara
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, 120 New Scotland Avenue, Albany, NY 12208, USA; Department of Biomedical Sciences, University at Albany School of Public Health, Albany, NY 12201, USA
| | - Anastasiya Yermakova
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, 120 New Scotland Avenue, Albany, NY 12208, USA; Department of Biomedical Sciences, University at Albany School of Public Health, Albany, NY 12201, USA
| | - Nicholas J. Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, 120 New Scotland Avenue, Albany, NY 12208, USA; Department of Biomedical Sciences, University at Albany School of Public Health, Albany, NY 12201, USA
| |
Collapse
|
18
|
Buttle DJ, Bramwell H, Hollander AP. Proteolytic mechanisms of cartilage breakdown: a target for arthritis therapy? Mol Pathol 2010; 48:M167-77. [PMID: 16696000 PMCID: PMC407956 DOI: 10.1136/mp.48.4.m167] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- D J Buttle
- Institute for Bone & Joint Medicine, Department of Human Metabolism & Clinical Biochemistry, University of Sheffield Medical School, Beech Hill Road, Sheffield S10 2RX
| | | | | |
Collapse
|
19
|
Puolakkainen M, Lee A, Nosaka T, Fukushi H, Kuo CC, Campbell LA. Retinoic acid inhibits the infectivity and growth of Chlamydia pneumoniae in epithelial and endothelial cells through different receptors. Microb Pathog 2007; 44:410-6. [PMID: 18162363 DOI: 10.1016/j.micpath.2007.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 11/06/2007] [Accepted: 11/09/2007] [Indexed: 10/22/2022]
Abstract
Chlamydia pneumoniae is a human respiratory pathogen that has also been associated with cardiovascular disease. C. pneumoniae infection accelerates atherosclerotic plaque development in hyperlipidemic animals and promotes oxidation of low density lipoprotein in vitro. All-trans-retinoic acid (ATRA), an antioxidant, has been shown to inhibit C. pneumoniae infectivity for endothelial cells by preventing binding of the organism to the M6P/IGF2 receptor on the cell surface. This current study investigates whether ATRA similarly affects C. pneumoniae infectivity of epithelial cells, which are the primary site of infection in the respiratory tract, and the effects on intracellular growth in both endothelial and epithelial cells. Because ATRA binds to both the nuclear retinoid acid receptor (RAR) and the M6P/IGF2 receptor, 4-[(E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)-1-propenyl]benzoic acid (TTNPB), an ATRA analog, which binds to the RAR but not the M6P/IGF2 receptor was used to differentiate the receptor mediating the effects of ATRA. The results of this study showed two separate effects of ATRA. The first effect is through interaction with the M6P/IGF2 receptor on the cell surface preventing attachment of the organism (inhibition by ATRA but not TTNPB) in endothelial cells and the second is through the nuclear receptor (inhibition by both ATRA and TTNPB) which inhibits growth in both epithelial and endothelial cells.
Collapse
Affiliation(s)
- Mirja Puolakkainen
- Department of Pathobiology, University of Washington, Box 357238, Seattle, WA 91895, USA
| | | | | | | | | | | |
Collapse
|
20
|
Sarkanen JR, Nykky J, Siikanen J, Selinummi J, Ylikomi T, Jalonen TO. Cholesterol supports the retinoic acid-induced synaptic vesicle formation in differentiating human SH-SY5Y neuroblastoma cells. J Neurochem 2007; 102:1941-1952. [PMID: 17540009 DOI: 10.1111/j.1471-4159.2007.04676.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synaptic vesicle formation, vesicle activation and exo/endocytosis in the pre-synaptic area are central steps in neuronal communication. The formation and localization of synaptic vesicles in human SH-SY5Y neuroblastoma cells, differentiated with 12-o-tetradecanoyl-phorbol-13-acetate, dibutyryl cyclic AMP, all-trans-retinoic acid (RA) and cholesterol, was studied by fluorescence microscopy and immunocytochemical methods. RA alone or together with cholesterol, produced significant neurite extension and formation of cell-to-cell contacts. Synaptic vesicle formation was followed by anti-synaptophysin (SypI) and AM1-43 staining. SypI was only weakly detected, mainly in cell somata, before 7 days in vitro, after which it was found in neurites. Depolarization of the differentiated cells with high potassium solution increased the number of fluorescent puncta, as well as SypI and AM1-43 co-localization. In addition to increase in the number of synaptic vesicles, RA and cholesterol also increased the number and distribution of lysosome-associated membrane protein 2 labeled lysosomes. RA-induced Golgi apparatus fragmentation was partly avoided by co-treatment with cholesterol. The SH-SY5Y neuroblastoma cell line, differentiated by RA and cholesterol and with good viability in culture, is a valuable tool for basic studies of neuronal metabolism, specifically as a model for dopaminergic neurons.
Collapse
Affiliation(s)
- Jertta-Riina Sarkanen
- Cell Research Center, Medical School, University of Tampere, Tampere, FinlandDivision of Biochemistry, Department of Biological and Environmental Science and NanoScience Center, University of Jyväskylä, Jyväskylä, FinlandInstitute of Signal Processing, Tampere University of Technology, Tampere, FinlandDepartment of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
| | - Jonna Nykky
- Cell Research Center, Medical School, University of Tampere, Tampere, FinlandDivision of Biochemistry, Department of Biological and Environmental Science and NanoScience Center, University of Jyväskylä, Jyväskylä, FinlandInstitute of Signal Processing, Tampere University of Technology, Tampere, FinlandDepartment of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
| | - Jutta Siikanen
- Cell Research Center, Medical School, University of Tampere, Tampere, FinlandDivision of Biochemistry, Department of Biological and Environmental Science and NanoScience Center, University of Jyväskylä, Jyväskylä, FinlandInstitute of Signal Processing, Tampere University of Technology, Tampere, FinlandDepartment of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
| | - Jyrki Selinummi
- Cell Research Center, Medical School, University of Tampere, Tampere, FinlandDivision of Biochemistry, Department of Biological and Environmental Science and NanoScience Center, University of Jyväskylä, Jyväskylä, FinlandInstitute of Signal Processing, Tampere University of Technology, Tampere, FinlandDepartment of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
| | - Timo Ylikomi
- Cell Research Center, Medical School, University of Tampere, Tampere, FinlandDivision of Biochemistry, Department of Biological and Environmental Science and NanoScience Center, University of Jyväskylä, Jyväskylä, FinlandInstitute of Signal Processing, Tampere University of Technology, Tampere, FinlandDepartment of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
| | - Tuula O Jalonen
- Cell Research Center, Medical School, University of Tampere, Tampere, FinlandDivision of Biochemistry, Department of Biological and Environmental Science and NanoScience Center, University of Jyväskylä, Jyväskylä, FinlandInstitute of Signal Processing, Tampere University of Technology, Tampere, FinlandDepartment of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
21
|
Bharadwaj S, Rathore SS, Ghosh PC. Enhancement of the cytotoxicity of liposomal ricin by the carboxylic ionophore monensin and the lysosomotropic amine NH4Cl in Chinese hamster ovary cells. Int J Toxicol 2006; 25:349-59. [PMID: 16940007 DOI: 10.1080/10915810600846195] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Ricin was encapsulated in negatively charged liposomes and its effect on the cytotoxicity was compared with native ricin in Chinese hamster ovarian (CHO) cells. The cytotoxicity of ricin, as measured by a marker protein synthesis (incorporation of 3H-leucine), was reduced markedly (300-fold) following encapsulation in liposomes. Lactose, a potent inhibitor of ricin cytotoxicity, had no effect on the binding, internalization, and cytotoxicity of liposomal ricin, indicating that liposomal ricin enter into mammalian cells by an alternative route, bypassing galactose-mediated endocytic pathway. Both monensin (a carboxylic ionophore) and NH4Cl (a lysosomotropic amine) markedly enhances the cytotoxicity of liposomal ricin, indicating endocytotic uptake of liposomal ricin. The degree of potentiation of the cytotoxicity of liposomal ricin by both monensin and NH4Cl was significantly higher (441- and 51-fold) as compared to native ricin (62.5- and 12.5-fold). The extent of exocytosis of free ricin was found to be much higher as compared to liposomal ricin; on the other hand, the extent of degradation of free and liposomal ricin was identical. Consequently, the intracellular level of liposomal ricin was increased to 3.5-fold. This higher level of intracellular liposomal ricin may allow more efficient ricin A-chain release into the cytosol under the influence of NH4Cl and monensin. Monensin-induced potentiation of liposomal ricin was prevented by brefeldin A, indicating that in the presence of monensin, the liposomal ricin was efficiently routed through the Golgi apparatus en route to the cytosol. Thus, liposomal ricin in combination with monensin may have potential application for selective elimination of malignant cells.
Collapse
Affiliation(s)
- Seemha Bharadwaj
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, India
| | | | | |
Collapse
|
22
|
Lu M, Gong X, Lu Y, Guo J, Wang C, Pan Y. Molecular Cloning and Functional Characterization of a Cell-permeable Superoxide Dismutase Targeted to Lung Adenocarcinoma Cells. J Biol Chem 2006; 281:13620-13627. [PMID: 16551617 DOI: 10.1074/jbc.m600523200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In clinical oncology, many trials with superoxide dismutase (SOD) have failed to demonstrate antitumor ability and in many cases even caused deleterious effects because of low tumor-targeting ability. In the current research, the Nostoc commune Fe-SOD coding sequence was amplified from genomic DNA. In addition, the single chain variable fragment (ScFv) was constructed from the cDNA of an LC-1 hybridoma cell line secreting anti-lung adenocarcinoma monoclonal antibody. After modification, the SOD and ScFv were fused and co-expressed, and the resulting fusion protein produced SOD and LC-1 antibody activity. Tracing SOD-ScFv by fluorescein isothiocyanate and superoxide anions (O2*-) in SPC-A-1 cells showed that the fusion protein could recognize and enter SPC-A-1 cells to eliminate O2*-. The lower oxidative stress resulting from the decrease in cellular O2*- delayed the cell cycle at G1 and significantly slowed SPC-A-1 cell growth in association with the dephosphorylation of the serine-threonine protein kinase Akt and expression of p27kip1. The tumor-targeting fusion protein resulting from this research overcomes two disadvantages of SODs previously used in the clinical setting, the inability to target tumor cells or permeate the cell membrane. These findings lay the groundwork for development of an efficient antitumor drug targeted by the ScFv.
Collapse
Affiliation(s)
- Min Lu
- Institute of Biochemistry, Zhejiang University, Hangzhou, 310027, China
| | - Xingguo Gong
- Institute of Biochemistry, Zhejiang University, Hangzhou, 310027, China.
| | - Yuwen Lu
- Institute of Biochemistry, Zhejiang University, Hangzhou, 310027, China
| | - Jianjun Guo
- Institute of Biochemistry, Zhejiang University, Hangzhou, 310027, China
| | - Chenhui Wang
- Institute of Biochemistry, Zhejiang University, Hangzhou, 310027, China
| | - Yuanjiang Pan
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou, 310027, China.
| |
Collapse
|
23
|
Tzankov A. Retinoic acid-induced Golgi apparatus disruption in F2000 fibroblasts: a model for enhanced intracellular retrograde transport. JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2003; 36:265-8. [PMID: 12787480 DOI: 10.5483/bmbrep.2003.36.3.265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Retinoic acid (RA) can transform the Golgi apparatus (GA) into a diffuse vacuolar aggregate and increase the toxicity of some immunotoxins that enter into cells by receptor-mediated endocytosis. An ultramorphological study of the RA-induced GA disruption was performed on F2000 fibroblasts. Cultures were treated with 0.11 to 30 microM RA for 7-180 min. The endocytosis of Limax flavus agglutinin-peroxidase conjugate (LFA), and the interactions between a phorbol ester (PMA) and RA concerning GA disruption, were examined. Exposure to 0.33 microM RA for 20 min transformed the GA into vacuolar aggregate. These vacuoles were not involved in endocytosis since they remained unstained after endocytosis of LFA. However, the lysosomes were involved in endocytosis, as they were strongly stained. Therefore, a RA-induced shift towards lysosomal routing of the entered LFA was presumed. Exposure to PMA made cells resistant to the Golgi-disturbing effects of RA, indicating that protein kinase C plays an important role in this process.
Collapse
Affiliation(s)
- Alexandar Tzankov
- Institute of Pathology, University of Innsbruck, Mullerstr. 44, A-6020 Innsbruck, Austria.
| |
Collapse
|
24
|
Abstract
Tadpoles of Bufo bufo at the end of premetamorphosis were exposed to the action of retinol diluted in the rearing water at two concentrations: 20 and 40 microM, for 48 h. Assuming that retinol causes growth suppression by cell death induction, the TUNEL reaction for the detection of apoptotic cells was performed on paraffin sections of pancreas. The results showed that retinol induced a significant dose-dependent increase of number of apoptotic cells with respect to controls, in which apoptosis was scarce. Electron microscopic observations of treated specimens showed that the effects of retinol mostly occurred on exocrine cells: condensed crescent-shaped nuclear chromatin, dark cytoplasm with long projections; conversely, mitochondria and endoplasmic reticulum appeared unaffected.
Collapse
Affiliation(s)
- F Accordi
- Dipartimento di Biologia Animale e dell'Uomo, Università La Sapienza, Roma, Italy.
| | | |
Collapse
|
25
|
Saxena SK, Sirdeshmukh R, Ardelt W, Mikulski SM, Shogen K, Youle RJ. Entry into cells and selective degradation of tRNAs by a cytotoxic member of the RNase A family. J Biol Chem 2002; 277:15142-6. [PMID: 11839736 DOI: 10.1074/jbc.m108115200] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Onconase (P-30 protein), an enzyme in the ribonuclease A superfamily, exerts cytostatic, cytotoxic, and antiviral activity when added to the medium of growing mammalian cells. We find that onconase enters living mammalian cells and selectively cleaves tRNA with no detectable degradation of rRNA. The RNA specificity of onconase in vitro using reticulocyte lysate and purified RNA substrates indicates that proteins associated with rRNA protect the rRNA from the onconase ribonucleolytic action contributing to the cellular tRNA selectivity of onconase. The onconase-mediated tRNA degradation in cells appears to be accompanied by increased levels of tRNA turnover and induction of tRNA synthesis perhaps in response to the selective toxin-induced loss of tRNA. Degradation products of tRNA(3)(Lys), which acts as a primer for HIV-1 replication, were clearly detected in cells infected with HIV-1 and treated with sublethal concentrations of onconase. However, a new synthesis of tRNA(3)(Lys) also seemed to occur in these cells resulting in plateauing of the steady-state levels of this tRNA. We conclude that the degradation of tRNAs may be a primary factor in the cytotoxic activity of onconase.
Collapse
Affiliation(s)
- Shailendra K Saxena
- Biochemistry Section, Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892-1414, USA
| | | | | | | | | | | |
Collapse
|
26
|
Ghidoni R, Sala G, Giuliani A. Use of sphingolipid analogs: benefits and risks. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1439:17-39. [PMID: 10395962 DOI: 10.1016/s1388-1981(99)00074-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- R Ghidoni
- INSERM U410, Neuroendocrinologie et Biologie Cellulaire Digestives, Faculté de Médecine Xavier Bichat, Paris, France.
| | | | | |
Collapse
|
27
|
Brodowicz T, Wiltschke C, Kandioler-Eckersberger D, Grunt TW, Rudas M, Schneider SM, Hejna M, Budinsky A, Zielinski CC. Inhibition of proliferation and induction of apoptosis in soft tissue sarcoma cells by interferon-alpha and retinoids. Br J Cancer 1999; 80:1350-8. [PMID: 10424735 PMCID: PMC2363080 DOI: 10.1038/sj.bjc.6690528] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Uncontrolled proliferation and a defect of apoptosis constitute crucial elements in the development and progression of tumours. Among many other biological response modifiers known to influence these mechanisms, the efficacy of retinoids and interferons in the treatment of various malignant entities is currently matter of discussion. In the present study, we have investigated the effects of 9-cis-retinoic acid (9cRA), 13-cis-retinoic acid (13cRA), all-trans-retinoic acid (tRA) and interferon-alpha on proliferation and apoptosis of human soft tissue sarcoma (STS) cell lines HTB-82 (rhabdomyosarcoma), HTB-91 (fibrosarcoma), HTB-92 (liposarcoma), HTB-93 (synovial sarcoma) and HTB-94 (chondrosarcoma) in relation to p53 genotype as well as p53 expression. HTB-91, HTB-92 and HTB-94 STS cells exhibited mutant p53, whereas wild-type p53 was found in HTB-93 STS cells, and a normal p53 status in HTB-82 STS cells, carrying a silent point mutation only. Interferon-alpha, irrespective of p53 status, inhibited the proliferation of all five cell lines dose- and time-dependently. Similarly, 9cRA, 13cRA and tRA decreased the proliferation of HTB-82 and HTB-93 STS cells, whereas the proliferation of p53-mutated HTB-91, HTB-92 and HTB-94 STS cells remained unchanged. Furthermore, only 9cRA and tRA were capable of inducing apoptosis in HTB-82 and HTB-93 STS cells, whereas HTB-91, HTB-92 and HTB-94 STS cells did not undergo apoptosis under the influence of 9cRA or tRA. Retinoic acid receptor (RAR)-alpha and RAR-beta mRNA were not detectable by Northern blot analysis in the five STS cell lines, whereas mRNA for the universal retinoic acid receptor, RAR-gamma, was expressed in all STS cell lines indicating that retinoid resistance was not associated with a lack of RAR expression. Apoptosis was not induced by interferon-alpha or 13cRA in any of the five STS cell lines tested. Our results indicate that within the panel of tested STS cell lines, inhibition of proliferation and induction of apoptosis result from different mechanisms which differ in their dependence upon the presence of intact p53.
Collapse
Affiliation(s)
- T Brodowicz
- Clinical Division of Oncology, Department of Medicine I, University Hospital, Vienna, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tada H, Sasada R, Kawaguchi Y, Kojima I, Gullick WJ, Salomon DS, Igarashi K, Seno M, Yamada H. Processing and juxtacrine activity of membrane-anchored betacellulin. J Cell Biochem 1999. [DOI: 10.1002/(sici)1097-4644(19990301)72:3<423::aid-jcb11>3.0.co;2-p] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
29
|
Abstract
We have studied the effect of several structurally related mansonones on the cytotoxicity of plant and bacterial toxins in Vero and BER-40, a brefeldin A-resistant mutant of Vero cells. Mansonone-D (MD), a sesquiterpenoid ortho-naphthoquinone, inhibited the cytotoxicity of ricin, modeccin, Pseudomonas toxin, and diphtheria toxin in Vero cells to different extents. The inhibition of ricin cytotoxicity was dose dependent and reversed upon removal of the drug. Protection of ricin cytotoxicity was also observed in the presence of cycloheximide, indicating that de novo protein synthesis is not required for the protective effect. Although MD inhibited the degradation and excretion of ricin, the binding and internalization of ricin was not affected. In contrast, MD strongly reduced the specific binding of diphtheria toxin in Vero cells. Fluorescence microscopic studies show that MD treatment dramatically alters the morphology of the Golgi apparatus in Vero cells. The kinetic studies reveal that the protection of ricin cytotoxicity is the consequence of decreased toxin translocation to the cytosol in MD-treated cells. The reactive ortho-quinone moiety of MD is important for the protective effect as thespesone, a para-naphthoquinone with a heterocyclic ring structure identical to that of MD, did not inhibit the cytotoxicity of toxins. Thespone, a dehydromansonone-D, lacking two hydrogens from the heterocyclic dihydrofuran ring of MD, inhibited the cytotoxicity of ricin, but was albeit less potent than MD. Neither mansonone-E nor mansonone-H with reactive ortho-quinone moiety, but with a different heterocyclic structure, had any effect on the cytotoxicity of ricin indicating that the protective effect of MD is specifically related to the overall structure of the metabolite.
Collapse
Affiliation(s)
- M P Nambiar
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
30
|
Shumakov VI, Maisjuk IG, Agapov II, Shamshiev AT, Muromtceva IB, Maluchenko NV, Pfueller U, Palmer RA, Lentzen H, Tonevitsky AG. In vitro efficacy of conjugates of anti-CD45 monoclonal antibodies with plant toxin A-chains. Transplant Proc 1998; 30:971-3. [PMID: 9636392 DOI: 10.1016/s0041-1345(98)00114-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- V I Shumakov
- Institute of Transplantology and Artificial Organs, Moscow, Russia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
One of the major discoveries of effective therapeutics is the use of targeted treatment, such as antibody-directed toxins, i.e. immunotoxins; however, this medicine delivery strategy is still at a developmental stage. A number of problems need to be resolved; one is their inefficacy when applied in vivo. Research has stimulated interest in this area through the use of chemical reagents and other moieties to increase the activity of immunotoxins. In this article, reagents that can potentiate the cytotoxicity of immunotoxins are reviewed and the mechanisms that increase activity of immunotoxins are discussed. Lysosomotropic amines, especially ammonium chloride and chloroquine, may raise the pH value of the lysosome in which the conjugates enter. Carboxylic ionophores, e.g. monensin, can influence Golgi vacuolation, which may facilitate the routing of conjugates, augmenting activity. Calcium channel antagonists may increase immunotoxin killing through morphological or other mechanisms that are not yet well understood. Viral particles and surface structure can enhance the cytotoxicity of conjugates, probably through the mechanism of disrupting endosomes. In addition, cytokines, beta-adrenergic blockers, immunosuppressive agents (cyclosporin A) and some antibiotics (daunorubicin) can be used to increase the effect of immunotoxins.
Collapse
Affiliation(s)
- M Wu
- Department of Biology, University of Leeds, UK
| |
Collapse
|
32
|
Taupiac MP, Alami M, Beaumelle B. Translocation of full-length Pseudomonas exotoxin from endosomes is driven by ATP hydrolysis but requires prior exposure to acidic pH. J Biol Chem 1996; 271:26170-3. [PMID: 8824263 DOI: 10.1074/jbc.271.42.26170] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We attached human transferrin to Pseudomonas exotoxin A (PE) to specifically localize this toxin to the endosomal compartment and study its translocation from purified endosomes using a cell-free assay. Transferrin was linked to PE via a disulfide bond. Chemical derivatization inactivated the PE cell-binding domain, and transferrin-PE was found to be endocytosed via the transferrin receptor only. Transferrin was also conjugated to a truncated PE with no receptor-binding domain (PE46). After labeling mouse lymphocytes with radiolabeled transferrin-PE or transferrin-PE46 and endosome isolation, selective translocation of the full-sized toxin portion of the conjugate was observed in a cell-free system. This translocation was strictly dependent upon ATP hydrolysis and was not affected when the acidity of the endosome lumen was neutralized using weak bases, protonophores, or bafilomycin A1. Nevertheless, when present during cell labeling, inhibitors of endosome acidification prevented PE from acquiring translocation competence. Similar inhibition was observed when endocytosis was performed in the presence of brefeldin A, a drug known to interfere with the delivery of endocytic tracers to acidic endosomes. Our data indicate that full-length PE can be transferred to the cytosol directly from endosomes during intoxication by PE conjugates and that, although exposure to acidic pH is a prerequisite for translocation, ATP hydrolysis directly provides the energy required for PE translocation.
Collapse
Affiliation(s)
- M P Taupiac
- UMR 5539 CNRS, Département Biologie-Santé, Université Montpellier II, 34095 Montpellier Cedex 05, France
| | | | | |
Collapse
|
33
|
Saxena SK, Gravell M, Wu YN, Mikulski SM, Shogen K, Ardelt W, Youle RJ. Inhibition of HIV-1 production and selective degradation of viral RNA by an amphibian ribonuclease. J Biol Chem 1996; 271:20783-8. [PMID: 8702832 DOI: 10.1074/jbc.271.34.20783] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Ribonucleases appear to have physiologic roles in host defense against cancer, viruses, and other parasites. Previously it was shown that select ribonucleases added to cells concurrently with virions blocked human immunodeficiency virus, type I (HIV-1) infection of H9 cells. We now report that a ribonuclease homologous to RNase A, named onconase, inhibits virus replication in chronically HIV-1-infected human cells without killing the virally infected cell. Examining the mechanism of this inhibition shows that onconase enters the infected cells and degrades HIV-1 RNA without degrading ribosomal RNA or the three different cellular messenger RNAs analyzed. The homologous human pancreatic RNase lacks anti-viral activity. Comparing recombinant forms of onconase and a onconase-human RNase chimera shows that the N-terminal 9 amino acids and the pyroglutamyl residue of onconase are required for full anti-viral activity. Thus extracellular ribonucleases can enter cells, metabolize select RNAs, and inhibit HIV virion production within viable replicating cells.
Collapse
Affiliation(s)
- S K Saxena
- Biochemistry Section of the Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Pincus SH, Wehrly K, Cole R, Fang H, Lewis GK, McClure J, Conley AJ, Wahren B, Posner MR, Notkins AL, Tilley SA, Pinter A, Eiden L, Teintze M, Dorward D, Tolstikov VV. In vitro effects of anti-HIV immunotoxins directed against multiple epitopes on HIV type 1 envelope glycoprotein 160. AIDS Res Hum Retroviruses 1996; 12:1041-51. [PMID: 8827220 DOI: 10.1089/aid.1996.12.1041] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We have used a panel of anti-gp160 MAbs to construct anti-HIV immunotoxins by coupling antibodies to ricin A chain (RAC). The ability of the immunotoxins to kill HIV-1-infected cells and halt the spread of infection was tested in tissue culture on persistently and acutely infected cell lines and primary lymphocyte cultures stimulated with phytohemagglutinin (PHA blasts). Laboratory strains and clinical isolates of HIV both were tested. The constitution and antigen-binding capacity of the immunotoxins were confirmed by ELISA and indirect immunofluorescence. Immunotoxins that bind epitopes exposed on the cell surface effectively killed persistently infected cells, although killing was not directly proportional to binding of immunotoxin to cell. The activity of anti-gp41, but not anti-gp120, immunotoxins was markedly enhanced in the presence of soluble CD4 or peptides corresponding to the CDR3 region of CD4. CD4-mediated enhancement of anti-gp41 immunotoxin activity was observed for laboratory strains neutralized by sCD4 and for clinical isolates that were resistant to neutralization by sCD4. Immunotoxin action was potentiated by brefeldin A, bafilomycin A1, cortisone, and an amphipathic fusion peptide, but not by cytochalasin D, nocodazol, monodansyl cadaverine, or trans-retinoic acid. Anti-HIV immunotoxins are useful tool with which to study the functional expression of gp120/gp41 antigens on the surface of HIV-infected cells, as well as potential AIDS therapeutics. Because these studies relate to the accessibility of viral antigens to antibody-mediated attack, these studies also have relevance for vaccine development.
Collapse
Affiliation(s)
- S H Pincus
- Laboratory of Microbial Structure and Function, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana 59840, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Wu Y, Saxena SK, Ardelt W, Gadina M, Mikulski SM, De Lorenzo C, D'Alessio G, Youle RJ. A study of the intracellular routing of cytotoxic ribonucleases. J Biol Chem 1995; 270:17476-81. [PMID: 7542240 DOI: 10.1074/jbc.270.29.17476] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Several ribonucleases serve as cytotoxic agents in host defense and in physiological cell death pathways. Although certain members of the pancreatic ribonuclease A superfamily can be toxic when applied to the outside of cells, they become thousands of times more toxic when artificially introduced into the cytosol, indicating that internalization is the rate-limiting step for cytotoxicity. We have used three agents that disrupt the Golgi apparatus by distinct mechanisms, retinoic acid, brefeldin A, and monensin, to probe the intracellular pathways ribonucleases take to reach the cytosol. Retinoic acid and monensin potentiate the cytotoxicity of bovine seminal RNase, Onconase, angiogenin, and human ribonuclease A 100 times or more. Retinoic acid-mediated potentiation of ribonucleases is completely blocked by brefeldin A. Ribonucleases appear to route more efficiently into the cytosol through the Golgi apparatus disrupted by monensin or retinoic acid. Intracellular RNA degradation by BS-RNase increased more than 100 times in the presence of retinoic acid confirming that the RNase reaches the cytosol and indicating that degradation of RNA is the intracellular lesion causing toxicity. As retinoic acid alone and Onconase are in clinical trials for cancer therapy, combinations of RNases and retinoic acid in vivo may offer new clinical utility.
Collapse
Affiliation(s)
- Y Wu
- Biochemistry Section, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Mohanraj D, Ramakrishnan S. Cytotoxic effects of ricin without an interchain disulfide bond: genetic modification and chemical crosslinking studies. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1243:399-406. [PMID: 7727515 DOI: 10.1016/0304-4165(94)00166-u] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Ricin is a toxic glycoprotein made of two polypeptide chains (A and B) linked by a disulfide bond. Ricin binds to cells by the B chain and is then internalized. The interchain disulfide bond is believed to be reduced in endosomes, and the A chain is then subsequently translocated to cytoplasm where it inactivates ribosomes. To understand the role of the disulfide bond in ricin toxicity, we prepared two types of ricin molecules. First, cysteine 259 of the A chain was mutated to an alanine residue. The mutant A chain was then reassociated with the native B chain to determine whether ricin is biologically active in the absence of an interchain disulfide bond. Reassociated mutant ricin showed a 40-fold reduction in biological activity. Binding studies using a hydrophobic fluorescence probe indicated that the associated complex was stable only at neutral pH and became highly unstable at a lower pH characteristic of the endosomal milieu. In the second construct, the interchain disulfide bond was replaced with a non-reducible bond by chemical derivatization. Interestingly, the non-reducible ricin molecule was equally cytotoxic as native ricin. These results show: (i) that the interchain disulfide bond is necessary to hold the A chain and the B chain together at endosomal pH, and (ii) that intact ricin may be transported to the cytoplasm where proteolysis or hydrolysis may occur to release the biologically active moiety.
Collapse
Affiliation(s)
- D Mohanraj
- Department of Pharmacology, University of Minnesota, Minneapolis 55455, USA
| | | |
Collapse
|
37
|
Redfern CP, Lovat PE, Malcolm AJ, Pearson AD. Gene expression and neuroblastoma cell differentiation in response to retinoic acid: differential effects of 9-cis and all-trans retinoic acid. Eur J Cancer 1995; 31A:486-94. [PMID: 7576951 DOI: 10.1016/0959-8049(95)00066-r] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Retinoic acid has considerable potential for the chemoprevention and chemotherapy of cancer. Neuroblastoma cells differentiate in response to retinoic acid in vitro, an observation that has led to clinical trials using either the 13-cis or all-trans isomers of retinoic acid. We review the effects of retinoic acid on neuroblastoma, and the potential involvement of nuclear retinoic acid receptors (RARs) and retinoid X receptors (RXRs). 9-cis retinoic acid is a ligand for RXRs, and we review recent data on the differential effects of 9-cis and all-trans retinoic acid on neuroblastoma differentiation and proliferation in vitro, and possible mechanisms of action via hetero- and homodimers of RARs and RXRs. Although there is uncertainty whether or not 9-cis retinoic acid produces its biological effects primarily via RXR homodimers, in vitro data suggest that this isomer of retinoic acid or stable analogues may have considerable potential for the treatment of resistant, disseminated neuroblastoma.
Collapse
Affiliation(s)
- C P Redfern
- Department of Medicine, Medical School, University of Newcastle, U.K
| | | | | | | |
Collapse
|