1
|
Lee SH, Khoo ASB, Griffiths JR, Mat Lazim N. Metabolic regulation of the tumour and its microenvironment: The role of Epstein-Barr virus. Int J Cancer 2025; 156:488-498. [PMID: 39291683 DOI: 10.1002/ijc.35192] [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: 07/15/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
The Epstein-Barr virus (EBV), the first identified human tumour virus, infects over 95% of the individuals globally and has the potential to induce different types of cancers. It is increasingly recognised that EBV infection not only alters cellular metabolism, contributing to neoplastic transformation, but also utilises several non-cell autonomous mechanisms to shape the metabolic milieu in the tumour microenvironment (TME) and its constituent stromal and immune cells. In this review, we explore how EBV modulates metabolism to shape the interactions between cancer cells, stromal cells, and immune cells within a hypoxic and acidic TME. We highlight how metabolites resulting from EBV infection act as paracrine factors to regulate the TME, and how targeting them can disrupt barriers to immunotherapy.
Collapse
Affiliation(s)
- Shen-Han Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Alan Soo-Beng Khoo
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - John R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Norhafiza Mat Lazim
- Department of Otorhinolaryngology-Head & Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
2
|
Marvi PK, Das P, Jafari A, Hassan S, Savoji H, Srinivasan S, Rajabzadeh AR. Multifunctional Carbon Dots In Situ Confined Hydrogel for Optical Communication, Drug Delivery, pH Sensing, Nanozymatic Activity, and UV Shielding Applications. Adv Healthc Mater 2025:e2403876. [PMID: 39757485 DOI: 10.1002/adhm.202403876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/29/2024] [Indexed: 01/07/2025]
Abstract
Inspired by the emerging potential of photoluminescent hydrogels, this work unlocks new avenues for advanced biosensing, bioimaging, and drug delivery applications. Carbon quantum dots (CDs) are deemed particularly promising among various optical dyes, for enhancing polymeric networks with superior physical and chemical properties. This study presents the synthesis of CDs derived from Prunella vulgaris, a natural plant resource, through a single-step hydrothermal process, followed by their uniform integration into hydrogel matrices via an in situ free radical graft polymerization. The resulting CD-integrated hydrogels exhibit multifunctionality in biomedical applications, featuring a diffusion-controlled drug release mechanism, permit concurrent delivery of photoluminescent CDs and therapeutic agents, enabling real-time monitoring over 32 h. In addition, these hydrogels function as a broad-range optical pH sensor (pH 3-11), provide robust ultraviolet (UV) shielding, and demonstrate nanozyme-like peroxidase activity. Critically, biocompatibility tests confirm their non-cytotoxicity toward fibroblast cells, establishing these hydrogels as promising candidates for diverse biomedical applications. These include advanced wound dressings that monitor the healing process and detect infection through pH sensing, and promote healing through the nanozymatic activity, all while maintaining a moist wound microenvironment. These hydrogels demonstrate exceptional suitability for advanced smart drug delivery, effective UV-blocking, and as innovative platforms for in vivo sensing and bioimaging.
Collapse
Affiliation(s)
- Parham Khoshbakht Marvi
- School of Biomedical Engineering, McMaster University, 1280 Main Street West Hamilton, Hamilton, Ontario, L8S 4L7, Canada
| | - Poushali Das
- School of Biomedical Engineering, McMaster University, 1280 Main Street West Hamilton, Hamilton, Ontario, L8S 4L7, Canada
| | - Arman Jafari
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
| | - Shiza Hassan
- School of Engineering Practice and Technology, McMaster University, 1280 Main Street West Hamilton, Hamilton, Ontario, L8S 4L7, Canada
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, QC, H3T 1J4, Canada
- Center for Applied Research On Polymers and Composites (CREPEC), Montreal, QC, H3A 0C3, Canada
| | - Seshasai Srinivasan
- School of Biomedical Engineering, McMaster University, 1280 Main Street West Hamilton, Hamilton, Ontario, L8S 4L7, Canada
- School of Engineering Practice and Technology, McMaster University, 1280 Main Street West Hamilton, Hamilton, Ontario, L8S 4L7, Canada
| | - Amin Reza Rajabzadeh
- School of Biomedical Engineering, McMaster University, 1280 Main Street West Hamilton, Hamilton, Ontario, L8S 4L7, Canada
- School of Engineering Practice and Technology, McMaster University, 1280 Main Street West Hamilton, Hamilton, Ontario, L8S 4L7, Canada
| |
Collapse
|
3
|
Frackowiak JE, Kubica P, Kosno M, Potęga A, Owczarek-Grzymkowska K, Borzyszkowska-Bukowska J, Laskowski T, Paluszkiewicz E, Mazerska Z. Distinct cellular uptake patterns of two anticancer unsymmetrical bisacridines and their metabolic transformation in tumor cells. J Pharm Biomed Anal 2025; 252:116493. [PMID: 39368137 DOI: 10.1016/j.jpba.2024.116493] [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: 08/05/2024] [Revised: 09/12/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
Unsymmetrical bisacridines (UAs) represent a novel class of anticancer agents. Their high cytotoxicity towards multiple human cancer cell lines and inhibition of human tumor xenograft growth in nude mice signal their potential for cancer treatment. Therefore, the mechanism of their strong biological activity is broadly investigated. Here, we explore the efflux and metabolism of UAs, as both strongly contribute to the development of drug resistance in cancer cells. We tested two highly cytotoxic UAs, C-2028 and C-2045, as well as their glucuronic acid and glutathione conjugates in human cancer cell lines (HepG2 and LS174T). As a point of reference for cell-based systems, we examined the rate of UA metabolic conversion in cell-free systems. A multiple reaction monitoring (MRM)-mass spectrometry (MS) method was developed in the present study for analysis of UAs and their metabolic conversion in complex biological matrices. Individual analytes were identified by several features: their retention time, mass-to-charge ratio and unique fragmentation pattern. The rate of UA uptake and metabolic transformation was monitored for 24 h in cell extracts and cell culture medium. Both UAs were rapidly internalized by cells. However, C-2028 was gradually accumulated, while C-2045 was eventually released from cells during treatment. UAs demonstrated limited metabolic conversion in cells. The glucuronic acid conjugate was excreted, whereas the glutathione conjugate was deposited in cancer cells. Our results obtained from cell-free and cell-based systems, using a uniform MRM-MS method, will provide valuable insight into the mechanism of UA biological activity in diverse biological models.
Collapse
Affiliation(s)
- Joanna E Frackowiak
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland.
| | - Paweł Kubica
- Department of Analytical Chemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland
| | - Michał Kosno
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland
| | - Agnieszka Potęga
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland
| | - Katarzyna Owczarek-Grzymkowska
- Department of Biochemistry, Bioanalytical Laboratory, Faculty of Medicine, Medical University of Gdańsk, 1 Dębinki Str., Gdańsk 80-211, Poland
| | - Julia Borzyszkowska-Bukowska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland
| | - Tomasz Laskowski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland
| | - Ewa Paluszkiewicz
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland
| | - Zofia Mazerska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland.
| |
Collapse
|
4
|
Icard P, Prieto M, Coquerel A, Fournel L, Gligorov J, Noel J, Mouren A, Dohan A, Alifano M, Simula L. Why and how citrate may sensitize malignant tumors to immunotherapy. Drug Resist Updat 2025; 78:101177. [PMID: 39612545 DOI: 10.1016/j.drup.2024.101177] [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: 12/19/2023] [Revised: 11/12/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Immunotherapy, either alone or in combination with chemotherapy, has demonstrated limited efficacy in a variety of solid cancers. Several factors contribute to explaining primary or secondary resistance. Among them, cancer cells, whose metabolism frequently relies on aerobic glycolysis, promote exhaustion of cytotoxic immune cells by diverting the glucose in the tumor microenvironment (TME) to their own profit, while secreting lactic acid that sustains the oxidative metabolism of immunosuppressive cells. Here, we propose to combine current treatment based on the use of immune checkpoint inhibitors (ICIs) with high doses of sodium citrate (SCT) because citrate inhibits cancer cell metabolism (by targeting both glycolysis and oxidative metabolism) and may active anti-tumor immune response. Indeed, as showed in preclinical studies, SCT reduces cancer cell growth, promoting cell death and chemotherapy effectiveness. Furthermore, since the plasma membrane citrate carrier pmCIC is mainly expressed in cancer cells and low or not expressed in immune and non-transformed cells, we argue that the inhibition of cancer cell metabolism by SCT may increase glucose availability in the TME, thus promoting functionality of anti-tumor immune cells. Concomitantly, the decrease in the amount of lactic acid in the TME may reduce the functionality of immunosuppressive cells. Preclinical studies have shown that SCT can enhance the anti-tumor immune response through an enhancement of T cell infiltration and activation, and a repolarization of macrophages towards a TAM1-like phenotype. Therefore, this simple and cheap strategy may have a major impact to increase the efficacy of current immunotherapies in human solid tumors and we encourage testing it in clinical trials.
Collapse
Affiliation(s)
- Philippe Icard
- INSERM U1086 ANTICIPE, Interdisciplinary Research Unit for Cancers Prevention and Treatment, BioTICLA Laboratory, Université de Caen Normandie, Caen, France; Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France.
| | - Mathilde Prieto
- Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France
| | - Antoine Coquerel
- INSERM U1075, COMETE « Mobilités: Attention, Orientation, Chronobiologie », Université Caen, France
| | - Ludovic Fournel
- Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France; INSERM UMR-S 1007, Cellular Homeostasis and Cancer, Paris-Descartes University, Paris
| | - Joseph Gligorov
- Oncology Department, Tenon Hospital, Pierre et Marie Curie University, Paris
| | - Johanna Noel
- Oncology Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France
| | - Adrien Mouren
- Département d'Innovation Thérapeutique et d´Essais Précoces (DITEP), Institut Gustave Roussy, Villejuif 94805, France
| | - Anthony Dohan
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris 75014, France; Radiology Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, Paris-Descartes University, Paris, France
| | - Luca Simula
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris 75014, France.
| |
Collapse
|
5
|
Berger KD, Puthenpeedikakkal AMK, Mathews DH, Fu D. Structural impact of 3-methylcytosine modification on the anticodon stem of a neuronally-enriched arginine tRNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624017. [PMID: 39605410 PMCID: PMC11601484 DOI: 10.1101/2024.11.18.624017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
All tRNAs undergo a series of chemical modifications to fold and function correctly. In mammals, the C32 nucleotide in the anticodon loop of tRNA-Arg-CCU and UCU is methylated to form 3-methylcytosine (m3C). Deficiency of m3C in arginine tRNAs has been linked to human neurodevelopmental disorders, indicating a critical biological role for m3C modification. However, the structural repercussions of m3C modification are not well understood. Here, we examine the structural effects of m3C32 modification on the anticodon stem loop (ASL) of human tRNA-Arg-UCU-4-1, a unique tRNA with enriched expression in the central nervous system. Optical melting experiments demonstrate that m3C modification can locally disrupt nearby base pairing within the ASL while simultaneously stabilizing the ASL electrostatically, resulting in little net change thermodynamically. The isoenergetic nature of the C32 - A38 pair vs the m3C32 - A38 pair may help discriminate against structures not adopting canonical C32 - A38 pairings, as most other m3C pairings are unfavorable. Furthermore, multidimensional NMR reveals that after m3C modification there are changes in hairpin loop structure and dynamics, the structure of A37, and the neighboring A31 - U39 base pair. However, these structural changes after modification are made while maintaining the shape of the C32 - A38 pairing, which is essential for efficient tRNA function in translation. These findings suggests that m3C32 modification could alter interactions of tRNA-Arg isodecoders with one or more binding partners while simultaneously maintaining the tRNA's ability to function in translation.
Collapse
Affiliation(s)
- Kyle D. Berger
- Department of Biology, Center for RNA Biology, University of Rochester, Rochester, New York 14627, United States
| | - Anees M. K. Puthenpeedikakkal
- Department of Biochemistry & Biophysics, Center for RNA Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - David H. Mathews
- Department of Biochemistry & Biophysics, Center for RNA Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Dragony Fu
- Department of Biology, Center for RNA Biology, University of Rochester, Rochester, New York 14627, United States
| |
Collapse
|
6
|
Verma A, Patel K, Kumar A. Targeting drug resistance in breast cancer: the potential of miRNA and nanotechnology-driven delivery systems. NANOSCALE ADVANCES 2024:d4na00660g. [PMID: 39569336 PMCID: PMC11575621 DOI: 10.1039/d4na00660g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Breast cancer is the second leading cause of cancer-related deaths in females worldwide. Despite significant advancements in treatment, drug resistance remains a major challenge, limiting the effectiveness of therapies and leading to dismal outcomes. Approximately 50% of HER2+ breast cancer patients develop resistance to trastuzumab, and patients with triple-negative breast cancer often experience resistance to first-line therapies. The drug resistance mechanisms involve altered drug uptake, enhanced DNA repair, and dysregulated apoptosis pathways. MicroRNAs are essential in regulating cellular processes involved in both homeostasis and disease. Recent data suggest that microRNAs can overcome drug resistance by regulating the pathways that confer drug resistance. Combining different conventional anticancer agents with microRNA therapies holds promise for enhancing treatment effectiveness against drug resistant breast cancer. Advancements in nano-drug delivery systems have facilitated the effective delivery of microRNAs by improving their stability, targeting specific cells, and enhancing cellular uptake. This review elucidates the recent advancements in microRNA-based therapies, their effects on gene expression, and their clinical efficacy in overcoming drug resistance in breast cancer.
Collapse
Affiliation(s)
- Aditi Verma
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| | - Krunal Patel
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| |
Collapse
|
7
|
Mari M, Boniburini M, Tosato M, Zanni F, Bonini F, Faglioni F, Cuoghi L, Belluti S, Imbriano C, Asti M, Ferrari E. Bridging pyrimidine hemicurcumin and Cisplatin: Synthesis, coordination chemistry, and in vitro activity assessment of a novel Pt(II) complex. J Inorg Biochem 2024; 260:112702. [PMID: 39163714 DOI: 10.1016/j.jinorgbio.2024.112702] [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: 06/28/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 08/22/2024]
Abstract
In the upcoming decades, the incidence and mortality rates of cancer are expected to rise globally, with colorectal and prostate cancers among the most prevalent types. Despite advancements in molecular targeted therapy, platinum-based chemotherapies remain the cornerstone of treatment, especially for colorectal and prostate cancer, with oxaliplatin and cisplatin being extremely effective due to their DNA-targeting capabilities. In our pursuit of new platinum-based chemotherapeutics that are potentially less toxic and more effective, we have explored the combination of the Pt-binding groups of the diaminocyclohexane ring used in oxaliplatin, with the stable amino-pyrimidine hemicurcumin moiety. This new derivative exhibit improved stability in physiological conditions and increased solubility in aqueous media, demonstrating promising effects on cell proliferation of both colorectal and prostate cells. We report herein the complete synthesis and chemical characterization in solution of the new derivative [(1R,2R)-N1-(3-(4-((E)-2-(2-Amino-6-methylpyrimidin-4-yl)vinyl)-2-methoxyphenoxy) propyl) cyclohexane-1,2-diamine] (MPYD). Our analysis includes an examination of its acid-base equilibria, speciation and stability in physiological conditions. The synthesis and in situ formation of Pt(II) complexes were investigated by nuclear magnetic resonance spectroscopy, while density functional theory calculations were employed to elucidate the chemical structure in solution. Results on the biological activity were obtained through cell viability assays on different colorectal and prostate cell lines (HCT116, HT29, PC3 and LNCaP).
Collapse
Affiliation(s)
- Matteo Mari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via Campi, 103, 41125 Modena, Italy
| | - Matteo Boniburini
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via Campi, 103, 41125 Modena, Italy
| | - Marianna Tosato
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via Campi, 103, 41125 Modena, Italy; Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, Azienda USL-IRCCS Reggio Emilia, via Amendola 2, 42122 Reggio Emilia, Italy
| | - Francesca Zanni
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via Campi, 103, 41125 Modena, Italy
| | - Filippo Bonini
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via Campi, 103, 41125 Modena, Italy
| | - Francesco Faglioni
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via Campi, 103, 41125 Modena, Italy
| | - Laura Cuoghi
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 213/d, 41125 Modena, Italy.
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 213/d, 41125 Modena, Italy.
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 213/d, 41125 Modena, Italy.
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, Azienda USL-IRCCS Reggio Emilia, via Amendola 2, 42122 Reggio Emilia, Italy.
| | - Erika Ferrari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via Campi, 103, 41125 Modena, Italy.
| |
Collapse
|
8
|
Ziental D, Czarczynska-Goslinska B, Wysocki M, Ptaszek M, Sobotta Ł. Advances and perspectives in use of semisolid formulations for photodynamic methods. Eur J Pharm Biopharm 2024; 204:114485. [PMID: 39255919 DOI: 10.1016/j.ejpb.2024.114485] [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: 06/12/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
Although nearly 30 years have passed since the introduction of the first clinically approved photosensitizer for photodynamic therapy, progress in developing new pharmaceutical formulations remains unsatisfactory. This review highlights that despite years of research, many recurring challenges and issues remain unresolved. The paper includes an analysis of selected essential studies involving aminolevulinic acid and its derivatives, as well as other photosensitizers with potential for development as medical products. Among various possible vehicles, special attention is given to gelatin, alginates, poly(ethylene oxide), polyacrylic acid, and chitosan. The focus is particularly on infectious and cancerous diseases. Key aspects of developing new semi-solid drug forms should prioritize the creation of easily manufacturable and biocompatible preparations for clinical use. At the same time, new formulations should preserve the primary function of photosensitizers, which is the generation of reactive oxygen species capable of destroying pathogenic cells or tumors. Additionally, the use of adjuvant properties of carriers, which can enhance the effectiveness of macrocycles, is emphasized, especially in chitosan-based antibacterial formulations. Current research indicates that many promising dyes and macrocyclic compounds with high potential as photosensitizers in photodynamic therapy remain unexplored in formulation and development work. This review outlines potential new and previously explored pathways for advancing photosensitizers as active pharmaceutical ingredients (APIs).
Collapse
Affiliation(s)
- Daniel Ziental
- Chair and Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland.
| | - Beata Czarczynska-Goslinska
- Chair and Department of Pharmaceutical Technology, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Marcin Wysocki
- Chair and Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Marcin Ptaszek
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Łukasz Sobotta
- Chair and Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| |
Collapse
|
9
|
Peng X, Liu Y, Peng F, Wang T, Cheng Z, Chen Q, Li M, Xu L, Man Y, Zhang Z, Tan Y, Liu Z. Aptamer-controlled stimuli-responsive drug release. Int J Biol Macromol 2024; 279:135353. [PMID: 39245104 DOI: 10.1016/j.ijbiomac.2024.135353] [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: 06/12/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Aptamers have been widely researched and applied in nanomedicine due to their programmable, activatable, and switchable properties. However, there are few reviews on aptamer-controlled stimuli-responsive drug delivery. This article highlights the mechanisms and advantages of aptamers in the construction of stimuli-responsive drug delivery systems. We summarize the assembly/reconfiguration mechanisms of aptamers in controlled release systems. The assembly and drug release strategies of drug delivery systems are illustrated. Specifically, we focus on the binding mechanisms to the target and the factors that induce/inhibit the binding to the stimuli, such as strand, pH, light, and temperature. The applications of aptamer-based stimuli-responsive drug release are elaborated. The challenges are discussed, and the future directions are proposed.
Collapse
Affiliation(s)
- Xingxing Peng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Feicheng Peng
- Hunan Institute for Drug Control, Changsha 410001, Hunan Province, PR China
| | - Ting Wang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhongyu Cheng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
10
|
Hsu CY, A Abbood M, Kadhim Abbood N, Hemid Al-Athari AJ, Shather AH, Talib Kareem A, Hassan Ahmed H, Yadav A. Mechanical quantum analysis on the role of transition metals on the delivery of metformin anticancer drug by the boron phosphide nanotube. Comput Methods Biomech Biomed Engin 2024; 27:1920-1930. [PMID: 37847195 DOI: 10.1080/10255842.2023.2267718] [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: 06/06/2023] [Revised: 09/12/2023] [Accepted: 09/30/2023] [Indexed: 10/18/2023]
Abstract
We scrutinized the impact of doping of X atoms (X = Fe, Co, Ni, Cu, and Zn) on the metformin (MF) drug delivery performance of a BP nanotube (BPNT) using density functional B3LYP calculations. The pristine BPNT was not ideal for the drug delivery of MF because of a weak interaction between the drug and nanotube. Doping of the Zn, Cu, Ni, Co, and Fe into the BPNT surface raised the adsorption energy of MF from -5.3 to -29.1, -28.7, -29.8, -32.1, and -26.9 kcal/mol, respectively, demonstrating that the sensitiveness of the metal-doped BPNT increased after increasing the radius atomic of metals. Ultimately, there was an increase in the adhesion performance and capacity of the MF after X (especially Co atom) doping, making the nanotube suitable for MF drug delivery. The mechanism of MF reaction with the BPNT changed from covalent bonding in the natural environment to hydrogen bonding in the cancerous cells with high acidity.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Manal A Abbood
- Division of Medical and Industrial Materials Science, Department of Applied Sciences, University of Technology, Iraq
| | - Nabeel Kadhim Abbood
- Chemical Engineering and Oil Refining Department, Basrah University for Oil and Gas, Oil and Gas Engineering College, Iraq
| | | | - A H Shather
- Department of Computer Engineering Technology, Al Kitab University, Altun Kopru, Kirkuk, Iraq
| | - Ashwaq Talib Kareem
- Collage of Pharmacy, National University of Science and Technology, Dhi Qa, Iraq
| | | | - Anupam Yadav
- Department of CEA, GLA University, Mathura, India
| |
Collapse
|
11
|
Tornetta MA, Whitaker BP, Cantwell OM, Pisors ED, Han L, MacWilliams MP, Jiang H, Zhou F, Chiu ML. The process using a synthetic library that generates multiple diverse human single domain antibodies. Antib Ther 2024; 7:283-294. [PMID: 39381136 PMCID: PMC11456836 DOI: 10.1093/abt/tbae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 10/10/2024] Open
Abstract
Background Single domain antibodies (sdAbs) possess unique characteristics that make them highly effective for developing complex therapeutics. Methods Our process uses a fully synthetic phage display library to generate single domain antibodies that can bind to disease relevant antigen conformations. A human IGHV3 family scaffold makes up the phage display libraries, and these VHO libraries are applied to diverse phage biopannings against target antigens. After NGS processing, unique VHOs undergo automated cloning into expression constructs followed by transfections and purifications. Binding assays were used to determine VHO binding behaviors to the target proteins. Additional VHO interactions are measured against endogenous targets on cells by way of flow cytometry, cell internalization, and activation assays. Results We show that a fully synthetic phage display library can generate VHOs that bind to disease relevant antigen conformations. The diverse biopanning methods and processing of next-generation sequencing generated many VHO paratopes. These different VHO sequences can be expressed as Fc fusion proteins. Various screening assays resulted in VHOs representing different epitopes or activities. During the hit evaluation, we demonstrate how screening can identify distinct VHO activities that have been used to generate differentiated drug molecules in various bispecific and multispecific antibody formats. Conclusion We demonstrate how screening can identify distinct VHO activities that have been used to generate differentiated drug molecules in various bispecific and multispecific antibody formats.
Collapse
Affiliation(s)
- Mark A Tornetta
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Brian P Whitaker
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Olivia M Cantwell
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Eileen D Pisors
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Lu Han
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| | - Maria P MacWilliams
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
| | - Hao Jiang
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| | - Fulai Zhou
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| | - Mark L Chiu
- Biologics Discovery Department, Tavotek Biotherapeutics, 727 Norristown Road, Spring house Innovation Park, Building 3, Suite 101, Lower Gywnedd, PA 19002, United States
- Biologics Discovery Department, Tavotek Biotherapeutics, Building C2, Suzhou Biomedical Industrial Park, Suzhou, Jiang Su 215000, China
| |
Collapse
|
12
|
Herrera-Ochoa D, Bravo I, Garzón-Ruiz A. Monitoring cancer treatments in melanoma cells using a fluorescence lifetime nanoprobe based on a CdSe/ZnS quantum dot functionalized with a peptide containing D-penicillamine and histidine. Colloids Surf B Biointerfaces 2024; 245:114265. [PMID: 39321721 DOI: 10.1016/j.colsurfb.2024.114265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Anticancer therapies with cisplatin and volasertib (BI-6727) were monitored by fluorescence lifetime imaging microscopy (FLIM) in live SK-Mel-2 melanoma cells. A CdSe/ZnS quantum dot functionalized with a peptide containing D-penicillamine and histidine (CdSe/ZnS-PH) was used as intracellular pH fluorescent probe. A faster cytosol acidification was observed for cells treated with cisplatin when compared to volasertib. The first changes in the intracellular pH were found after 2 hours of treatment with cisplatin and 8 hours with volasertib. Additionally, the relationship between cytosol acidification and apoptosis was investigated using an innovative methodology based on time-resolved fluorescence measurements. Similar low percentages of apoptotic cells were observed after short incubation periods (2 - 8 hours) with both drugs. In contrast, late apoptosis and death were found for a large fraction of cells during 24-hour incubation with cisplatin but not volasertib. Thus, the early acidification observed in cisplatin treatment could accelerate apoptosis and cell death. Despite volasertib treatment shows slower mechanism of action than cisplatin, similar inhibitory effects were found for both drugs at longer incubation periods (72 hours). This study proves the potential of CdSe/ZnS-PH nanoparticle as a fluorescence lifetime probe in the study of the mechanism of action of antitumor drugs.
Collapse
Affiliation(s)
- Diego Herrera-Ochoa
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, Albacete 02071, Spain
| | - Iván Bravo
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, Albacete 02071, Spain; Centro Regional de Investigaciones Biomédicas (CRIB), Unidad Asociada de Biomedicina (UCLM-CSIC), C/ Almansa, 14, Albacete 02008, Spain
| | - Andrés Garzón-Ruiz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, Albacete 02071, Spain.
| |
Collapse
|
13
|
Tan NK, Chan H, Lu Z, Zreiqat H, Lakhwani G, Lesani P, New EJ. Ultrasensitive Dual Fluorophore-Conjugated Carbon Dots for Intracellular pH Sensing in 3D Tumor Models. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47303-47313. [PMID: 39215383 DOI: 10.1021/acsami.4c10836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The dysregulation of pH has been linked to the onset of chronic conditions, such as cancer and neurological diseases. Consequently, the development of a highly sensitive tool for intracellular pH sensing is imperative to investigate the interplay between pH and the biochemical changes accompanying disease pathogenesis. Here, we present the development of a ratiometric fluorescent nanoprobe, NpRhoDot, designed for precisely measuring pH levels. We demonstrate its efficacy in sensitively reporting intracellular pH in monolayer A549 lung cancer cells, primary fibroblast cells, and 3D tumor spheroids derived from the DLD-1 colorectal adenocarcinoma cell line. NpRhoDot leverages a novel design, where stable carbon dots are functionalized with a pH-responsive ratiometric fluorescent probe comprising a naphthalimide-rhodamine moiety, NpRho1. This design confers NpRhoDot with the high pH sensitivity characteristics of organic fluorescent probes, along with excellent photostability up to 1 h and biocompatibility of carbon dots. Through one-photon and two-photon fluorescence microscopy, we validate the reliability of NpRhoDot for biosensing intracellular pH in monolayer and three-dimensional tumor models from pH 4 to 7.
Collapse
Affiliation(s)
- Nian Kee Tan
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hazel Chan
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Zufu Lu
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hala Zreiqat
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
| | - Girish Lakhwani
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence in Exciton Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pooria Lesani
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States of America
| | - Elizabeth J New
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
14
|
Chaudhuri A, Eijsink LE, Simeth NA. Optical control of pH via chromoselective photodosimetry. Chem Commun (Camb) 2024; 60:9745-9748. [PMID: 39171354 DOI: 10.1039/d4cc03076a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
The dynamic regulation of pH via an external stimulus is an attractive technique to gate chemical transformations. Applying photons of different energy, we preferentially address either a photoacid or a photobase donor in the same solutions and thus, present a technique to regulate the pH of a solution through light pulses.
Collapse
Affiliation(s)
- Amrita Chaudhuri
- Institute for Organic and Biomolecular Chemistry, Department of Chemistry, University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany.
| | - Linda E Eijsink
- Institute for Organic and Biomolecular Chemistry, Department of Chemistry, University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Nadja A Simeth
- Institute for Organic and Biomolecular Chemistry, Department of Chemistry, University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
15
|
Mohanan S, Guan X, Liang M, Karakoti A, Vinu A. Stimuli-Responsive Silica Silanol Conjugates: Strategic Nanoarchitectonics in Targeted Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301113. [PMID: 36967548 DOI: 10.1002/smll.202301113] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The design of novel drug delivery systems is exceptionally critical in disease treatments. Among the existing drug delivery systems, mesoporous silica nanoparticles (MSNs) have shown profuse promise owing to their structural stability, tunable morphologies/sizes, and ability to load different payload chemistry. Significantly, the presence of surface silanol groups enables functionalization with relevant drugs, imaging, and targeting agents, promoting their utility and popularity among researchers. Stimuli-responsive silanol conjugates have been developed as a novel, more effective way to conjugate, deliver, and release therapeutic drugs on demand and precisely to the selected location. Therefore, it is urgent to summarize the current understanding and the surface silanols' role in making MSN a versatile drug delivery platform. This review provides an analytical understanding of the surface silanols, chemistry, identification methods, and their property-performance correlation. The chemistry involved in converting surface silanols to a stimuli-responsive silica delivery system by endogenous/exogenous stimuli, including pH, redox potential, temperature, and hypoxia, is discussed in depth. Different chemistries for converting surface silanols to stimuli-responsive bonds are discussed in the context of drug delivery. The critical discussion is culminated by outlining the challenges in identifying silanols' role and overcoming the limitations in synthesizing stimuli-responsive mesoporous silica-based drug delivery systems.
Collapse
Affiliation(s)
- Shan Mohanan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Xinwei Guan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajay Karakoti
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajayan Vinu
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| |
Collapse
|
16
|
Koirala M, DiPaola M. Overcoming Cancer Resistance: Strategies and Modalities for Effective Treatment. Biomedicines 2024; 12:1801. [PMID: 39200265 PMCID: PMC11351918 DOI: 10.3390/biomedicines12081801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Resistance to cancer drugs is a complex phenomenon that poses a significant challenge in the treatment of various malignancies. This review comprehensively explores cancer resistance mechanisms and discusses emerging strategies and modalities to overcome this obstacle. Many factors contribute to cancer resistance, including genetic mutations, activation of alternative signaling pathways, and alterations in the tumor microenvironment. Innovative approaches, such as targeted protein degradation, immunotherapy combinations, precision medicine, and novel drug delivery systems, hold promise for improving treatment outcomes. Understanding the intricacies of cancer resistance and leveraging innovative modalities are essential for advancing cancer therapy.
Collapse
|
17
|
Timimi L, Wrobel AG, Chiduza GN, Maslen SL, Torres-Méndez A, Montaner B, Davis C, Minckley T, Hole KL, Serio A, Devine MJ, Skehel JM, Rubinstein JL, Schreiber A, Beale R. The V-ATPase/ATG16L1 axis is controlled by the V 1H subunit. Mol Cell 2024; 84:2966-2983.e9. [PMID: 39089251 DOI: 10.1016/j.molcel.2024.07.003] [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: 12/18/2023] [Revised: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 08/03/2024]
Abstract
Defects in organellar acidification indicate compromised or infected compartments. Recruitment of the autophagy-related ATG16L1 complex to pathologically neutralized organelles targets ubiquitin-like ATG8 molecules to perturbed membranes. How this process is coupled to proton gradient disruption is unclear. Here, we reveal that the V1H subunit of the vacuolar ATPase (V-ATPase) proton pump binds directly to ATG16L1. The V1H/ATG16L1 interaction only occurs within fully assembled V-ATPases, allowing ATG16L1 recruitment to be coupled to increased V-ATPase assembly following organelle neutralization. Cells lacking V1H fail to target ATG8s during influenza infection or after activation of the immune receptor stimulator of interferon genes (STING). We identify a loop within V1H that mediates ATG16L1 binding. A neuronal V1H isoform lacks this loop and is associated with attenuated ATG8 targeting in response to ionophores in primary murine and human iPSC-derived neurons. Thus, V1H controls ATG16L1 recruitment following proton gradient dissipation, suggesting that the V-ATPase acts as a cell-intrinsic damage sensor.
Collapse
Affiliation(s)
- Lewis Timimi
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Division of Medicine, University College London, London WC1E 6JF, UK
| | - Antoni G Wrobel
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - George N Chiduza
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Sarah L Maslen
- Proteomics STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Antonio Torres-Méndez
- Neural Circuits & Evolution Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Beatriz Montaner
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Colin Davis
- Cellular Degradation Systems Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Taylor Minckley
- Neural Circuit Bioengineering and Disease Modelling Laboratory, The Francis Crick Institute, London NW1 1AT, UK; UK Dementia Research Institute at King's College London, London SE5 9RX, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London SE5 9RX, UK
| | - Katriona L Hole
- Mitochondrial Neurobiology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Andrea Serio
- Neural Circuit Bioengineering and Disease Modelling Laboratory, The Francis Crick Institute, London NW1 1AT, UK; UK Dementia Research Institute at King's College London, London SE5 9RX, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London SE5 9RX, UK
| | - Michael J Devine
- Mitochondrial Neurobiology Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - J Mark Skehel
- Proteomics STP, The Francis Crick Institute, London NW1 1AT, UK
| | - John L Rubinstein
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, The University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medical Biophysics, The University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Anne Schreiber
- Cellular Degradation Systems Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Division of Medicine, University College London, London WC1E 6JF, UK.
| |
Collapse
|
18
|
Chen X, Wu D, Chen Z. Biomedical applications of stimuli-responsive nanomaterials. MedComm (Beijing) 2024; 5:e643. [PMID: 39036340 PMCID: PMC11260173 DOI: 10.1002/mco2.643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Nanomaterials have aroused great interests in drug delivery due to their nanoscale structure, facile modifiability, and multifunctional physicochemical properties. Currently, stimuli-responsive nanomaterials that can respond to endogenous or exogenous stimulus display strong potentials in biomedical applications. In comparison with conventional nanomaterials, stimuli-responsive nanomaterials can improve therapeutic efficiency and reduce the toxicity of drugs toward normal tissues through specific targeting and on-demand drug release at pathological sites. In this review, we summarize the responsive mechanism of a variety of stimulus, including pH, redox, and enzymes within pathological microenvironment, as well as exogenous stimulus such as thermal effect, magnetic field, light, and ultrasound. After that, biomedical applications (e.g., drug delivery, imaging, and theranostics) of stimuli-responsive nanomaterials in a diverse array of common diseases, including cardiovascular diseases, cancer, neurological disorders, inflammation, and bacterial infection, are presented and discussed. Finally, the remaining challenges and outlooks of future research directions for the biomedical applications of stimuli-responsive nanomaterials are also discussed. We hope that this review can provide valuable guidance for developing stimuli-responsive nanomaterials and accelerate their biomedical applications in diseases diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| |
Collapse
|
19
|
Yang K, Wu Z, Zhang K, Weir MD, Xu HHK, Cheng L, Huang X, Zhou W. Unlocking the potential of stimuli-responsive biomaterials for bone regeneration. Front Pharmacol 2024; 15:1437457. [PMID: 39144636 PMCID: PMC11322102 DOI: 10.3389/fphar.2024.1437457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
Bone defects caused by tumors, osteoarthritis, and osteoporosis attract great attention. Because of outstanding biocompatibility, osteogenesis promotion, and less secondary infection incidence ratio, stimuli-responsive biomaterials are increasingly used to manage this issue. These biomaterials respond to certain stimuli, changing their mechanical properties, shape, or drug release rate accordingly. Thereafter, the activated materials exert instructive or triggering effects on cells and tissues, match the properties of the original bone tissues, establish tight connection with ambient hard tissue, and provide suitable mechanical strength. In this review, basic definitions of different categories of stimuli-responsive biomaterials are presented. Moreover, possible mechanisms, advanced studies, and pros and cons of each classification are discussed and analyzed. This review aims to provide an outlook on the future developments in stimuli-responsive biomaterials.
Collapse
Affiliation(s)
- Ke Yang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhuoshu Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Keke Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Michael D. Weir
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Hockin H. K. Xu
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China School of Stomatology & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
20
|
Chembukavu SN, Lindsay AJ. Therapy-induced senescence in breast cancer: an overview. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:902-920. [PMID: 39280248 PMCID: PMC11390292 DOI: 10.37349/etat.2024.00254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/06/2024] [Indexed: 09/18/2024] Open
Abstract
Outcomes for women with breast cancer have improved dramatically in recent decades. However, many patients present with intrinsic drug resistance and others are initially sensitive to anti-cancer drugs but acquire resistance during the course of their treatment, leading to recurrence and/or metastasis. Drug therapy-induced senescence (TIS) is a form of drug resistance characterised by the induction of cell cycle arrest and the emergence of a senescence-associated secretory phenotype (SASP) that can develop in response to chemo- and targeted- therapies. A wide range of anticancer interventions can lead to cell cycle arrest and SASP induction, by inducing genotoxic stress, hyperactivation of signalling pathways or oxidative stress. TIS can be anti-tumorigenic in the short-term, but pro-tumorigenic in the long-term by creating a pro-inflammatory and immunosuppressive microenvironment. Moreover, the SASP can promote angiogenesis and epithelial-mesenchymal transition in neighbouring cells. In this review, we will describe the characteristics of TIS in breast cancer and detail the changes in phenotype that accompany its induction. We also discuss strategies for targeting senescent cancer cells in order to prevent or delay tumour recurrence.
Collapse
Affiliation(s)
- Suraj Narayanan Chembukavu
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| | - Andrew J Lindsay
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| |
Collapse
|
21
|
Prasad R, Jyothi VGS, Kommineni N, Bulusu RT, Mendes B, Lovell JF, Conde J. Biomimetic Ghost Nanomedicine-Based Optotheranostics for Cancer. NANO LETTERS 2024; 24:8217-8231. [PMID: 38848540 PMCID: PMC11247544 DOI: 10.1021/acs.nanolett.4c01534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Theranostic medicine combines diagnostics and therapeutics, focusing on solid tumors at minimal doses. Optically activated photosensitizers are significant examples owing to their photophysical and chemical properties. Several optotheranostics have been tested that convert light to imaging signals, therapeutic radicals, and heat. Upon light exposure, conjugated photosensitizers kill tumor cells by producing reactive oxygen species and heat or by releasing cancer antigens. Despite clinical trials, these molecularly conjugated photosensitizers require protection from their surroundings and a localized direction for site-specific delivery during blood circulation. Therefore, cell membrane biomimetic ghosts have been proposed for precise and safe delivery of these optically active large molecules, which are clinically relevant because of their biocompatibility, long circulation time, bypass of immune cell recognition, and targeting ability. This review focuses on the role of biomimetic nanoparticles in the treatment and diagnosis of tumors through light-mediated diagnostics and therapy, providing insights into their preclinical and clinical status.
Collapse
Affiliation(s)
- Rajendra Prasad
- School
of Biochemical Engineering, Indian Institute
of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Vaskuri G. S. Jyothi
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center (UTHSC), Memphis, Tennessee 38163, United States
| | - Nagavendra Kommineni
- Center
for Biomedical Research, Population Council, New York, New York 10065, United States
| | - Ravi Teja Bulusu
- Department
of Pharmaceutical Sciences, Florida A&M
University, Tallahassee, Florida 32307, United States
| | - Bárbara
B. Mendes
- NOVA
Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon 1169-056, Portugal
- ToxOmics,
NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, 1169-056, Portugal
| | - Jonathan F. Lovell
- Department
of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| | - João Conde
- NOVA
Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon 1169-056, Portugal
- ToxOmics,
NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, 1169-056, Portugal
| |
Collapse
|
22
|
Chisanga M, Masson JF. Machine Learning-Driven SERS Nanoendoscopy and Optophysiology. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:313-338. [PMID: 38701442 DOI: 10.1146/annurev-anchem-061622-012448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
A frontier of analytical sciences is centered on the continuous measurement of molecules in or near cells, tissues, or organs, within the biological context in situ, where the molecular-level information is indicative of health status, therapeutic efficacy, and fundamental biochemical function of the host. Following the completion of the Human Genome Project, current research aims to link genes to functions of an organism and investigate how the environment modulates functional properties of organisms. New analytical methods have been developed to detect chemical changes with high spatial and temporal resolution, including minimally invasive surface-enhanced Raman scattering (SERS) nanofibers using the principles of endoscopy (SERS nanoendoscopy) or optical physiology (SERS optophysiology). Given the large spectral data sets generated from these experiments, SERS nanoendoscopy and optophysiology benefit from advances in data science and machine learning to extract chemical information from complex vibrational spectra measured by SERS. This review highlights new opportunities for intracellular, extracellular, and in vivo chemical measurements arising from the combination of SERS nanosensing and machine learning.
Collapse
Affiliation(s)
- Malama Chisanga
- Département de Chimie, Institut Courtois, Quebec Center for Advanced Materials, Regroupement Québécois sur les Matériaux de Pointe, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage, Université de Montréal, Montréal, Québec, Canada;
| | - Jean-Francois Masson
- Département de Chimie, Institut Courtois, Quebec Center for Advanced Materials, Regroupement Québécois sur les Matériaux de Pointe, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage, Université de Montréal, Montréal, Québec, Canada;
| |
Collapse
|
23
|
Tverdokhlebova A, Sterin I, Jayaweera TM, Darie CC, Katz E, Smutok O. Chemical modification of α-chymotrypsin enabling its release from alginate hydrogel by electrochemically generated local pH change. Int J Biol Macromol 2024; 273:133234. [PMID: 38897525 DOI: 10.1016/j.ijbiomac.2024.133234] [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: 01/31/2024] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
This study investigates the controlled release of α-chymotrypsin from an alginate hydrogel matrix. When protein molecules entrapped in the hydrogel matrix have a size smaller than the hydrogel pores, their hold/release from the polymer matrix are controlled by the electrostatic interaction between the guest molecules and host polymer. α-Chymotrypsin, as a model protein, was chemically modified with negatively charged species to change its pI and to convert its attractive interaction with a negatively charged alginate hydrogel matrix to a repulsion interaction allowing its release by pH-triggered signal. Then, bulk pH changes and electrochemically controlled local pH changes resulting from oxygen reduction were used for the controlled release of the enzyme from the alginate hydrogel. Three batches of modified α-chymotrypsin with different linker/enzyme ratios were synthesized, and their release profiles were investigated. The activity of both unmodified and modified α-chymotrypsin was evaluated using a UV-visible spectrophotometer following the standard procedure for the enzymatic assay of α-chymotrypsin (EC 3.4.21.1) and compared across all batches. Direct infusion electrospray ionization mass spectrometry (DI ESI-MS) was used to analyze the protein modifications and their impact on the isoelectric point values.
Collapse
Affiliation(s)
- Anna Tverdokhlebova
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Ilya Sterin
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Taniya M Jayaweera
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Costel C Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Evgeny Katz
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA.
| | - Oleh Smutok
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA.
| |
Collapse
|
24
|
Wang XF, Duan YF, Zhu YQ, Liu ZJ, Wu YC, Liu TH, Zhang L, Wei JF, Liu GC. An Insulin-Modified pH-Responsive Nanopipette Based on Ion Current Rectification. SENSORS (BASEL, SWITZERLAND) 2024; 24:4264. [PMID: 39001043 PMCID: PMC11244478 DOI: 10.3390/s24134264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024]
Abstract
The properties of nanopipettes largely rely on the materials introduced onto their inner walls, which allow for a vast extension of their sensing capabilities. The challenge of simultaneously enhancing the sensitivity and selectivity of nanopipettes for pH sensing remains, hindering their practical applications. Herein, we report insulin-modified nanopipettes with excellent pH response performances, which were prepared by introducing insulin onto their inner walls via a two-step reaction involving silanization and amidation. The pH response intensity based on ion current rectification was significantly enhanced by approximately 4.29 times when utilizing insulin-modified nanopipettes compared with bare ones, demonstrating a linear response within the pH range of 2.50 to 7.80. In addition, insulin-modified nanopipettes featured good reversibility and selectivity. The modification processes were monitored using the I-V curves, and the relevant mechanisms were discussed. The effects of solution pH and insulin concentration on the modification results were investigated to achieve optimal insulin introduction. This study showed that the pH response behavior of nanopipettes can be greatly improved by introducing versatile molecules onto the inner walls, thereby contributing to the development and utilization of pH-responsive nanopipettes.
Collapse
Affiliation(s)
- Xu-Fan Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou 221004, China;
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou 221004, China
| | - Yi-Fan Duan
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou 221004, China;
| | - Yue-Qian Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou 221004, China;
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou 221004, China
| | - Zi-Jing Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou 221004, China
| | - Yu-Chen Wu
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou 221004, China;
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou 221004, China
| | - Tian-Hao Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou 221004, China;
| | - Ling Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
| | - Jian-Feng Wei
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Guo-Chang Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, China; (X.-F.W.); (Y.-F.D.); (Y.-Q.Z.); (Z.-J.L.); (T.-H.L.); (L.Z.)
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
25
|
Aventaggiato M, Arcangeli T, Vernucci E, Barreca F, Sansone L, Pellegrini L, Pontemezzo E, Valente S, Fioravanti R, Russo MA, Mai A, Tafani M. Pharmacological Activation of SIRT3 Modulates the Response of Cancer Cells to Acidic pH. Pharmaceuticals (Basel) 2024; 17:810. [PMID: 38931477 DOI: 10.3390/ph17060810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer cells modulate their metabolism, creating an acidic microenvironment that, in turn, can favor tumor progression and chemotherapy resistance. Tumor cells adopt strategies to survive a drop in extracellular pH (pHe). In the present manuscript, we investigated the contribution of mitochondrial sirtuin 3 (SIRT3) to the adaptation and survival of cancer cells to a low pHe. SIRT3-overexpressing and silenced breast cancer cells MDA-MB-231 and human embryonic kidney HEK293 cells were grown in buffered and unbuffered media at pH 7.4 and 6.8 for different times. mRNA expression of SIRT3 and CAVB, was measured by RT-PCR. Protein expression of SIRT3, CAVB and autophagy proteins was estimated by western blot. SIRT3-CAVB interaction was determined by immunoprecipitation and proximity ligation assays (PLA). Induction of autophagy was studied by western blot and TEM. SIRT3 overexpression increases the survival of both cell lines. Moreover, we demonstrated that SIRT3 controls intracellular pH (pHi) through the regulation of mitochondrial carbonic anhydrase VB (CAVB). Interestingly, we obtained similar results by using MC2791, a new SIRT3 activator. Our results point to the possibility of modulating SIRT3 to decrease the response and resistance of tumor cells to the acidic microenvironment and ameliorate the effectiveness of anticancer therapy.
Collapse
Affiliation(s)
- Michele Aventaggiato
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Tania Arcangeli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Enza Vernucci
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Federica Barreca
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Luigi Sansone
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Rome, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Laura Pellegrini
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Elena Pontemezzo
- European Hospital, New Fertility Group, Center for Reproductive Medicine, Via Portuense 700, 00149 Rome, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Rossella Fioravanti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Matteo Antonio Russo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Rome, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
26
|
Kromer C, Katz A, Feldmann I, Laux P, Luch A, Tschiche HR. A targeted fluorescent nanosensor for ratiometric pH sensing at the cell surface. Sci Rep 2024; 14:12302. [PMID: 38811698 PMCID: PMC11137054 DOI: 10.1038/s41598-024-62976-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024] Open
Abstract
The correlation between altered extracellular pH and various pathological conditions, including cancer, inflammation and metabolic disorders, is well known. Bulk pH measurements cannot report the extracellular pH value at the cell surface. However, there is a limited number of suitable tools for measuring the extracellular pH of cells with high spatial resolution, and none of them are commonly used in laboratories around the world. In this study, a versatile ratiometric nanosensor for the measurement of extracellular pH was developed. The nanosensor consists of biocompatible polystyrene nanoparticles loaded with the pH-inert reference dye Nile red and is surface functionalized with a pH-responsive fluorescein dye. Equipped with a targeting moiety, the nanosensor can adhere to cell membranes, allowing direct measurement of extracellular pH at the cell surface. The nanosensor exhibits a sensitive ratiometric pH response within the range of 5.5-9.0, with a calculated pKa of 7.47. This range optimally covers the extracellular pH (pHe) of most healthy cells and cells in which the pHe is abnormal, such as cancer cells. In combination with the nanosensors ability to target cell membranes, its high robustness, reversibility and its biocompatibility, the pHe nanosensor proves to be well suited for in-situ measurement of extracellular pH, even over extended time periods. This pH nanosensor has the potential to advance biomedical research by improving our understanding of cellular microenvironments, where extracellular pH plays an important role.
Collapse
Affiliation(s)
- Charlotte Kromer
- Product Materials and Nanotechnology, Department Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany.
| | - Aaron Katz
- Product Materials and Nanotechnology, Department Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Ines Feldmann
- Material-Microbiome Interactions, Department Materials and the Environment, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Peter Laux
- Product Materials and Nanotechnology, Department Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Andreas Luch
- Product Materials and Nanotechnology, Department Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Harald R Tschiche
- Product Materials and Nanotechnology, Department Chemical and Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| |
Collapse
|
27
|
Chávez JC, Carrasquel-Martínez G, Hernández-Garduño S, Matamoros Volante A, Treviño CL, Nishigaki T, Darszon A. Cytosolic and Acrosomal pH Regulation in Mammalian Sperm. Cells 2024; 13:865. [PMID: 38786087 PMCID: PMC11120249 DOI: 10.3390/cells13100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
As in most cells, intracellular pH regulation is fundamental for sperm physiology. Key sperm functions like swimming, maturation, and a unique exocytotic process, the acrosome reaction, necessary for gamete fusion, are deeply influenced by pH. Sperm pH regulation, both intracellularly and within organelles such as the acrosome, requires a coordinated interplay of various transporters and channels, ensuring that this cell is primed for fertilization. Consistent with the pivotal importance of pH regulation in mammalian sperm physiology, several of its unique transporters are dependent on cytosolic pH. Examples include the Ca2+ channel CatSper and the K+ channel Slo3. The absence of these channels leads to male infertility. This review outlines the main transport elements involved in pH regulation, including cytosolic and acrosomal pH, that participate in these complex functions. We present a glimpse of how these transporters are regulated and how distinct sets of them are orchestrated to allow sperm to fertilize the egg. Much research is needed to begin to envision the complete set of players and the choreography of how cytosolic and organellar pH are regulated in each sperm function.
Collapse
Affiliation(s)
- Julio C. Chávez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Gabriela Carrasquel-Martínez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
- CITMER, Medicina Reproductiva, México City 11520, Mexico
| | - Sandra Hernández-Garduño
- Departamento de Morfología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), México City 04510, Mexico;
| | - Arturo Matamoros Volante
- Department of Electrical and Computer Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Claudia L. Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Takuya Nishigaki
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| |
Collapse
|
28
|
Hashemzadeh T, Christofferson AJ, White KF, Barnard PJ. Experimental and theoretical studies of pH-responsive iridium(III) complexes of azole and N-heterocyclic carbene ligands. Dalton Trans 2024; 53:8478-8493. [PMID: 38687288 DOI: 10.1039/d3dt03766e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
A series of nine luminescent iridium(III) complexes with pH-responsive imidazole and benzimidazole ligands have been prepared and characterized. The first series of complexes were of the form [Ir(ppy)2(N^N)]+ or [Ir(ppy)2(C^N)]+ (where ppy is 2-phenylpyridine and N^N is 2-(2-pyridyl)imidazole or 2-(2-pyridyl)benzimidazole and C^N represents a pyridyl-triazolylidene-based N-heterocyclic carbene ligand). For these complexes, the benzimidazole group was either unsubstituted or substituted with electron-withdrawing (Cl) or electron-donating (Me) groups. The second series of complexes were of the form [Ir(phbim)2(N^N)]+ or [Ir(phbim)2(C^N)]+ (where phbim is 2-phenylbenzimidazole and N^N is either 2,2'-bipyridine or 1,10-phenanthroline and C^N is either a pyridyl-imidazolylidene or pyridyl-triazolylidene N-heterocyclic carbene ligand). UV-visible and photoluminescence pH titration studies showed that changing the protonation state of these complexes results in significant changes in the photoluminescence emission properties. The pKa values of prepared complexes were estimated from the spectroscopic pH titration data and these values show that the nature of the pH-sensitive ligands (either main or ancillary ligands) resulted in a significant capacity to modulate the pKa values for these compounds with values ranging from 5.19-11.22. Theoretical investigations into the nature of the electronic transitions for the different protonation states of compounds were performed and the results were consistent with the experimental results.
Collapse
Affiliation(s)
- Tahmineh Hashemzadeh
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia.
| | - Andrew J Christofferson
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
- ARC Centre of Excellence in Exciton Science, School of Science, RMIT University, Melbourne, Victoria 3001, Australia
| | - Keith F White
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia.
| | - Peter J Barnard
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia.
| |
Collapse
|
29
|
White B, Swietach P. What can we learn about acid-base transporters in cancer from studying somatic mutations in their genes? Pflugers Arch 2024; 476:673-688. [PMID: 37999800 PMCID: PMC11006749 DOI: 10.1007/s00424-023-02876-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023]
Abstract
Acidosis is a chemical signature of the tumour microenvironment that challenges intracellular pH homeostasis. The orchestrated activity of acid-base transporters of the solute-linked carrier (SLC) family is critical for removing the end-products of fermentative metabolism (lactate/H+) and maintaining a favourably alkaline cytoplasm. Given the critical role of pH homeostasis in enabling cellular activities, mutations in relevant SLC genes may impact the oncogenic process, emerging as negatively or positively selected, or as driver or passenger mutations. To address this, we performed a pan-cancer analysis of The Cancer Genome Atlas simple nucleotide variation data for acid/base-transporting SLCs (ABT-SLCs). Somatic mutation patterns of monocarboxylate transporters (MCTs) were consistent with their proposed essentiality in facilitating lactate/H+ efflux. Among all cancers, tumours of uterine corpus endometrial cancer carried more ABT-SLC somatic mutations than expected from median tumour mutation burden. Among these, somatic mutations in SLC4A3 had features consistent with meaningful consequences on cellular fitness. Definitive evidence for ABT-SLCs as 'cancer essential' or 'driver genes' will have to consider microenvironmental context in genomic sequencing because bulk approaches are insensitive to pH heterogeneity within tumours. Moreover, genomic analyses must be validated with phenotypic outcomes (i.e. SLC-carried flux) to appreciate the opportunities for targeting acid-base transport in cancers.
Collapse
Affiliation(s)
- Bobby White
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| |
Collapse
|
30
|
Chen H, Zhu MZ, Wang XT, Ai M, Li SS, Wan MY, Wang PY, Cai WW, Hou B, Xu F, Lang F, Qiu LY, Zhou YT. 1,25(OH) 2 D 3 inhibits Lewis lung cancer cell migration via NHE1-sensitive metabolic reprograming. IUBMB Life 2024; 76:182-199. [PMID: 37921568 DOI: 10.1002/iub.2789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023]
Abstract
High prevalence and metastasis rates are characteristics of lung cancer. Glycolysis provides energy for the development and metastasis of cancer cells. The 1,25-dihydroxy vitamin D3 (1,25(OH)2 D3 ) has been linked to reducing cancer risk and regulates various physiological functions. We hypothesized that 1,25(OH)2 D3 could be associated with the expression and activity of Na+ /H+ exchanger isoform 1 (NHE1) of Lewis lung cancer cells, thus regulating glycolysis as well as migration by actin reorganization. Followed by online public data analysis, Vitamin D3 receptor, the receptor of 1,25(OH)2 D3 has been proved to be abundant in lung cancers. We demonstrated that 1,25(OH)2 D3 treatment suppressed transcript levels, protein levels, and activity of NHE1 in LLC cells. Furthermore, 1,25(OH)2 D3 treatment resets the metabolic balance between glycolysis and OXPHOS, mainly including reducing glycolytic enzymes expression and lactate production. In vivo experiments showed the inhibition effects on tumor growth as well. Therefore, we concluded that 1,25(OH)2 D3 could amend the NHE1 function, which leads to metabolic reprogramming and cytoskeleton reconstruction, finally inhibits the cell migration.
Collapse
Affiliation(s)
- Hong Chen
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Mei-Zhen Zhu
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Xi-Ting Wang
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Min Ai
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
- Laboratory Animal Center of Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Shuang-Shuang Li
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
- Shanghai Seventh People's Hospital, Shanghai, People's Republic of China
| | - Ming-Yu Wan
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Pei-Yao Wang
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Wei-Wei Cai
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Bao Hou
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Fei Xu
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Li-Ying Qiu
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| | - Yue-Tao Zhou
- Wuxi Medical School, Jiangnan University, Wuxi, People's Republic of China
| |
Collapse
|
31
|
GAO HUAN, ZHANG JIE, KLEIJN TONYG, WU ZHAOYONG, LIU BING, MA YUJIN, DING BAOYUE, YIN DONGFENG. Dual ligand-targeted Pluronic P123 polymeric micelles enhance the therapeutic effect of breast cancer with bone metastases. Oncol Res 2024; 32:769-784. [PMID: 38560569 PMCID: PMC10972726 DOI: 10.32604/or.2023.044276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/13/2023] [Indexed: 04/04/2024] Open
Abstract
Bone metastasis secondary to breast cancer negatively impacts patient quality of life and survival. The treatment of bone metastases is challenging since many anticancer drugs are not effectively delivered to the bone to exert a therapeutic effect. To improve the treatment efficacy, we developed Pluronic P123 (P123)-based polymeric micelles dually decorated with alendronate (ALN) and cancer-specific phage protein DMPGTVLP (DP-8) for targeted drug delivery to breast cancer bone metastases. Doxorubicin (DOX) was selected as the anticancer drug and was encapsulated into the hydrophobic core of the micelles with a high drug loading capacity (3.44%). The DOX-loaded polymeric micelles were spherical, 123 nm in diameter on average, and exhibited a narrow size distribution. The in vitro experiments demonstrated that a pH decrease from 7.4 to 5.0 markedly accelerated DOX release. The micelles were well internalized by cultured breast cancer cells and the cell death rate of micelle-treated breast cancer cells was increased compared to that of free DOX-treated cells. Rapid binding of the micelles to hydroxyapatite (HA) microparticles indicated their high affinity for bone. P123-ALN/DP-8@DOX inhibited tumor growth and reduced bone resorption in a 3D cancer bone metastasis model. In vivo experiments using a breast cancer bone metastasis nude model demonstrated increased accumulation of the micelles in the tumor region and considerable antitumor activity with no organ-specific histological damage and minimal systemic toxicity. In conclusion, our study provided strong evidence that these pH-sensitive dual ligand-targeted polymeric micelles may be a successful treatment strategy for breast cancer bone metastasis.
Collapse
Affiliation(s)
- HUAN GAO
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
- Department of Pharmacy, The General Hospital of Xinjiang Military Region, Urumqi, 830000, China
| | - JIE ZHANG
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - TONY G. KLEIJN
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
- Department of Pathology, Laboratory of Experimental Oncology, Erasmus MC, Rotterdam, 3015 GD, The Netherlands
| | - ZHAOYONG WU
- Department of Pharmacy, Jiaxing Maternal and Child Health Care Hospital, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, China
| | - BING LIU
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
- Qinghai Enlu Biotechnology Co., Ltd., Haidong, 810700, China
| | - YUJIN MA
- Qinghai Enlu Biotechnology Co., Ltd., Haidong, 810700, China
| | - BAOYUE DING
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - DONGFENG YIN
- Department of Pharmacy, The General Hospital of Xinjiang Military Region, Urumqi, 830000, China
| |
Collapse
|
32
|
Reshetnyak YK, Andreev OA, Engelman DM. Aiming the magic bullet: targeted delivery of imaging and therapeutic agents to solid tumors by pHLIP peptides. Front Pharmacol 2024; 15:1355893. [PMID: 38545547 PMCID: PMC10965573 DOI: 10.3389/fphar.2024.1355893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 11/11/2024] Open
Abstract
The family of pH (Low) Insertion Peptides (pHLIP) comprises a tumor-agnostic technology that uses the low pH (or high acidity) at the surfaces of cells within the tumor microenvironment (TME) as a targeted biomarker. pHLIPs can be used for extracellular and intracellular delivery of a variety of imaging and therapeutic payloads. Unlike therapeutic delivery targeted to specific receptors on the surfaces of particular cells, pHLIP targets cancer, stromal and some immune cells all at once. Since the TME exhibits complex cellular crosstalk interactions, simultaneous targeting and delivery to different cell types leads to a significant synergistic effect for many agents. pHLIPs can also be positioned on the surfaces of various nanoparticles (NPs) for the targeted intracellular delivery of encapsulated payloads. The pHLIP technology is currently advancing in pre-clinical and clinical applications for tumor imaging and treatment.
Collapse
Affiliation(s)
- Yana K. Reshetnyak
- Physics Department, University of Rhode Island, Kingston, RI, United States
| | - Oleg A. Andreev
- Physics Department, University of Rhode Island, Kingston, RI, United States
| | - Donald M. Engelman
- Molecular Biophysics and Biochemistry Department, Yale, New Haven, CT, United States
| |
Collapse
|
33
|
Ma J, Lu X, Hao M, Wang Y, Guo Y, Wang Z. Real-time visualization the pH fluctuations of living cells with a ratiometric near-infrared fluorescent probe. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 306:123572. [PMID: 37922853 DOI: 10.1016/j.saa.2023.123572] [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: 09/04/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 11/07/2023]
Abstract
In situ real-time quantitative monitoring pH fluctuation in complex living systems is vitally significant. In the current work, a ratiometric near-infrared (NIR) probe (MCyOH) was developed to confront this challenge. MCyOH exhibited good sensitivity, photostability, reversibility, and an ideal pKa (pKa = 6.65). Ratiometric character of MCyOH is beneficial to accuracy detect the pH fluctuations in living cells under different stimulation. The observations showed that intracellular pH was decreased when HepG2 cells under oxidative stress or starvation conditions. In particular, HepG2 cells was acidulated after addition of ethanol, however, the acidification phenomenon was attenuated or disappeared when HepG2 cells preincubated with disulfiram or fomepizole. Finally, MCyOH was successfully applied to observe the increasement of intracellular pH when HepG2 cells treated with fomepizole individually. Overall, MCyOH would be a practical candidate to explore pH-associated physiological and pathological varieties.
Collapse
Affiliation(s)
- Jianlong Ma
- Department of Chemistry, Changzhi University, Changzhi 046011, PR China; Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, Gansu 730000, PR China
| | - Xiaofeng Lu
- Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, Gansu 730000, PR China
| | - Mingyao Hao
- Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, Gansu 730000, PR China; University of Chinese Academy of Sciences, 100049 Beijing, PR China
| | - Yumeng Wang
- Department of Chemistry, Changzhi University, Changzhi 046011, PR China
| | - Yong Guo
- Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, Gansu 730000, PR China.
| | - Zhijun Wang
- Department of Chemistry, Changzhi University, Changzhi 046011, PR China.
| |
Collapse
|
34
|
Tantawy MN, McIntyre JO, Yull F, Calcutt MW, Koktysh DS, Wilson AJ, Zu Z, Nyman J, Rhoades J, Peterson TE, Colvin D, McCawley LJ, Rook JM, Fingleton B, Crispens MA, Alvarez RD, Gore JC. Tumor therapy by targeting extracellular hydroxyapatite using novel drugs: A paradigm shift. Cancer Med 2024; 13:e6812. [PMID: 38239047 PMCID: PMC11025459 DOI: 10.1002/cam4.6812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND It has been shown that tumor microenvironment (TME) hydroxyapatite (HAP) is typically associated with many malignancies and plays a role in tumor progression and growth. Additionally, acidosis in the TME has been reported to play a key role in selecting for a more aggressive tumor phenotype, drug resistance and desensitization to immunotherapy for many types of cancers. TME-HAP is an attractive target for tumor detection and treatment development since HAP is generally absent from normal soft tissue. We provide strong evidence that dissolution of hydroxyapatite (HAP) within the tumor microenvironment (TME-HAP) using a novel therapeutic can be used to kill cancer cells both in vitro and in vivo with minimal adverse effects. METHODS We developed an injectable cation exchange nano particulate sulfonated polystyrene solution (NSPS) that we engineered to dissolve TME-HAP, inducing localized acute alkalosis and inhibition of tumor growth and glucose metabolism. This was evaluated in cell culture using 4T1, MDA-MB-231 triple negative breast cancer cells, MCF10 normal breast cells, and H292 lung cancer cells, and in vivo using orthotopic mouse models of cancer that contained detectable microenvironment HAP including breast (MMTV-Neu, 4T1, and MDA-MB-231), prostate (PC3) and colon (HCA7) cancer using 18 F-NaF for HAP and 18 F-FDG for glucose metabolism with PET imaging. On the other hand, H292 lung tumor cells that lacked detectable microenvironment HAP and MCF10a normal breast cells that do not produce HAP served as negative controls. Tumor microenvironment pH levels following injection of NSPS were evaluated via Chemical Exchange Saturation (CEST) MRI and via ex vivo methods. RESULTS Within 24 h of adding the small concentration of 1X of NSPS (~7 μM), we observed significant tumor cell death (~ 10%, p < 0.05) in 4T1 and MDA-MB-231 cell cultures that contain HAP but ⟨2% in H292 and MCF10a cells that lack detectable HAP and in controls. Using CEST MRI, we found extracellular pH (pHe) in the 4T1 breast tumors, located in the mammary fat pad, to increase by nearly 10% from baseline before gradually receding back to baseline during the first hour post NSPS administration. in the tumors that contained TME-HAP in mouse models, MMTV-Neu, 4T1, and MDA-MB-231, PC3, and HCA7, there was a significant reduction (p<0.05) in 18 F-Na Fuptake post NSPS treatment as expected; 18 F- uptake in the tumor = 3.8 ± 0.5 %ID/g (percent of the injected dose per gram) at baseline compared to 1.8 ±0.5 %ID/g following one-time treatment with 100 mg/kg NSPS. Of similar importance, is that 18 F-FDG uptake in the tumors was reduced by more than 75% compared to baseline within 24 h of treatment with one-time NSPS which persisted for at least one week. Additionally, tumor growth was significantly slower (p < 0.05) in the mice treated with one-time NSPS. Toxicity showed no evidence of any adverse effects, a finding attributed to the absence of HAP in normal soft tissue and to our therapeutic NSPS having limited penetration to access HAP within skeletal bone. CONCLUSION Dissolution of TME-HAP using our novel NSPS has the potential to provide a new treatment paradigm to enhance the management of cancer patients with poor prognosis.
Collapse
Affiliation(s)
- Mohammed N. Tantawy
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
- Departments of Radiology and Radiological SciencesVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - J. Oliver McIntyre
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
- Departments of Radiology and Radiological SciencesVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
- Department of PharmacologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Fiona Yull
- Department of PharmacologyVanderbilt UniversityNashvilleTennesseeUSA
- Department of Obstetrics and GynecologyVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - M. Wade Calcutt
- Department of BiochemistryVanderbilt UniversityNashvilleTennesseeUSA
- Mass Spectrometry Research Center of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
| | - Dmitry S. Koktysh
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Vanderbilt Institute of Nanoscale Science and EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Andrew J. Wilson
- Department of Obstetrics and GynecologyVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - Zhongliang Zu
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
- Departments of Radiology and Radiological SciencesVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - Jeff Nyman
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTennesseeUSA
- Orthopaedic SurgeryVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - Julie Rhoades
- Orthopaedic SurgeryVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
- Department of Veterans Affairs, Tennessee Valley Healthcare SystemNashvilleTennesseeUSA
| | - Todd E. Peterson
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
- Departments of Radiology and Radiological SciencesVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - Daniel Colvin
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Lisa J. McCawley
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Jerri. M. Rook
- Department of PharmacologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Barbara Fingleton
- Department of PharmacologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Marta Ann Crispens
- Department of Obstetrics and GynecologyVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
- Division of Gynecologic OncologyVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - Ronald D. Alvarez
- Department of Obstetrics and GynecologyVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| | - John C. Gore
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
- Departments of Radiology and Radiological SciencesVanderbilt Univerity Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
35
|
Sodhi H, Panitch A. A Tunable Glycosaminoglycan-Peptide Nanoparticle Platform for the Protection of Therapeutic Peptides. Pharmaceutics 2024; 16:173. [PMID: 38399234 PMCID: PMC10892384 DOI: 10.3390/pharmaceutics16020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The popularity of Glycosaminoglycans (GAGs) in drug delivery systems has grown as their innate ability to sequester and release charged molecules makes them adept in the controlled release of therapeutics. However, peptide therapeutics have been relegated to synthetic, polymeric systems, despite their high specificity and efficacy as therapeutics because they are rapidly degraded in vivo when not encapsulated. We present a GAG-based nanoparticle system for the easy encapsulation of cationic peptides, which offers control over particle diameter, peptide release behavior, and swelling behavior, as well as protection from proteolytic degradation, using a singular, organic polymer and no covalent linkages. These nanoparticles can encapsulate cargo with a particle diameter range spanning 130-220 nm and can be tuned to release cargo over a pH range of 4.5 to neutral through the modulation of the degree of sulfation and the molecular weight of the GAG. This particle system also confers better in vitro performance than the unencapsulated peptide via protection from enzymatic degradation. This method provides a facile way to protect therapeutic peptides via the inclusion of the presented binding sequence and can likely be expanded to larger, more diverse cargo as well, abrogating the complexity of previously demonstrated systems while offering broader tunability.
Collapse
Affiliation(s)
- Harkanwalpreet Sodhi
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| |
Collapse
|
36
|
Shirbhate E, Singh V, Kore R, Vishwakarma S, Veerasamy R, Tiwari AK, Rajak H. The Role of Cytokines in Activation of Tumour-promoting Pathways and Emergence of Cancer Drug Resistance. Curr Top Med Chem 2024; 24:523-540. [PMID: 38258788 DOI: 10.2174/0115680266284527240118041129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024]
Abstract
Scientists are constantly researching and launching potential chemotherapeutic agents as an irreplaceable weapon to fight the battle against cancer. Despite remarkable advancement over the past several decades to wipe out cancer through early diagnosis, proper prevention, and timely treatment, cancer is not ready to give up and leave the battleground. It continuously tries to find some other way to give a tough fight for its survival, either by escaping from the effect of chemotherapeutic drugs or utilising its own chemical messengers like cytokines to ensure resistance. Cytokines play a significant role in cancer cell growth and progression, and the present article highlights their substantial contribution to mechanisms of resistance toward therapeutic drugs. Multiple clinical studies have even described the importance of specific cytokines released from cancer cells as well as stromal cells in conferring resistance. Herein, we discuss the different mechanism behind drug resistance and the crosstalk between tumor development and cytokines release and their contribution to showing resistance towards chemotherapeutics. As a part of this review, different approaches to cytokines profile have been identified and employed to successfully target new evolving mechanisms of resistance and their possible treatment options.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, 495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, 495 009, (C.G.), India
| | - Rakesh Kore
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, 495 009, (C.G.), India
| | - Subham Vishwakarma
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, 495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100, Bedong, Kedah Darul Aman, Malaysia
| | - Amit K Tiwari
- Cancer & System Therapeutics, UAMS College of Pharmacy, UAMS - University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, 495 009, (C.G.) India
| |
Collapse
|
37
|
Siva M, Das K, Guha S, Sivagnanam S, Das G, Saha A, Stewart A, Maity B, Das P. Liposomes Containing Zinc-Based Chemotherapeutic Drug Block Proliferation and Trigger Apoptosis in Breast Cancer Cells. ACS APPLIED BIO MATERIALS 2023; 6:5310-5323. [PMID: 37988654 DOI: 10.1021/acsabm.3c00587] [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] [Indexed: 11/23/2023]
Abstract
Platinum-based chemotherapeutic drugs are effective in killing malignant cells but often trigger drug resistance or off-target side effects. Unlike platinum, zinc is used as an endogenous cofactor for several cellular enzymes and may, thus, display increased biocompatibility. In this present study, we have rationally designed and synthesized two substituted phenanthro[9,10-d]imidazole-based ligands L1 and L2 with pyridine and quinoline substitution at the 2 position and their corresponding Zn(II) complexes; (L1)2Zn and (L2)2Zn, which are characterized by standard analytical and spectroscopic methods. (L2)2Zn, but not (L1)2Zn has intrinsic fluorescence, indicating its potential utility in imaging applications. To facilitate cellular uptake, we generated liposomal formations with a phospholipid DMPC (1,2-Dimyristoyl-sn-glycero-3-phosphocholine) through molecular self-assembly. These liposomal formulations Lip-(L1)2Zn and Lip-(L2)2Zn were able to enter breast cancer cells, induce DNA fragmentation, arrest the cell cycle at the G0/G1 phase, decrease proliferation, and promote apoptosis by activating the DNA damage response. Importantly, both Lip-(L1)2Zn and Lip-(L2)2Zn decreased the size of breast cancer cell-based spheroids, indicating they may be capable of suppressing tumor growth. Our work represents an important proof-of-concept exercise demonstrating that successful liposomal formation of phenanthro[9,10-d]imidazole-based Zn(II) complexes with inherent optical properties have great promise for the development of imaging probes and efficient anticancer drugs.
Collapse
Affiliation(s)
- Mallayasamy Siva
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Kiran Das
- Department of Systems Biology, Centre of Biomedical Research (CBMR), SGPGI campus, Raebareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Subhabrata Guha
- Department of Signal Transduction and Biogenic Amines, Chittaranajan National Cancer Institute, 37, S.P.Mukherjee Road, Kolkata, West Bengal 700 026, India
| | - Subramaniyam Sivagnanam
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Gaurav Das
- Department of Signal Transduction and Biogenic Amines, Chittaranajan National Cancer Institute, 37, S.P.Mukherjee Road, Kolkata, West Bengal 700 026, India
| | - Abhijit Saha
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida 33458, United States
| | - Biswanath Maity
- Department of Systems Biology, Centre of Biomedical Research (CBMR), SGPGI campus, Raebareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Priyadip Das
- Department of Chemistry, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Tamil Nadu 603203, India
| |
Collapse
|
38
|
Yang X, Zhang C, Song M, Zhang Z, Zhou J, Zhang H, Ding Y. Enzyme-Silenced Nanosponges Prolong Intratumoral Lifetime to Facilitate Intercellular Relay Drug Delivery and Treatment Efficacy. ACS NANO 2023; 17:23568-23583. [PMID: 37976418 DOI: 10.1021/acsnano.3c06544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
The clinical application of nanomedicines faces the dilemma of improved safety but restricted efficacy due to the poor intratumoral bioavailability of chemotherapeutics. We here design an enzyme-silenced nanosponge that shares a long-term lifespan to reversibly exhale/inhale doxorubicin (DOX) for continuous intercellular relay delivery and improved intratumoral retention. The nanosponge is composed of a cationic lipid overlaying a hyaluronic acid derivative polyampholyte core for enveloping of DOX and hyaluronidase-1-targeted siRNA (siHyal1), and a lipoprotein shell decorated with fusion peptide 4F-tLyP-1 that was fused with apolipoprotein A-I (apoA-I) mimetic peptide 4F and tLyP-1 for tumor homing and extravasation into the tumor interstitium. Triggered by the intra/intercellular pH variation, the nanosponge core could reversibly swell in endo/lysosome (pH 5.0) for DOX release. Owing to the deprotonation, the nanosponge core shrinks back in cytoplasm (pH 7.4) for DOX reloading and continues the behavior after being secreted to the extracellular matrix (pH 6.8) via Golgi apparatus, which dramatically improves intratumoral DOX retention and availability. Concurrently, the intratumoral lifespan of the nanosponge is prolonged by siHyal1-specific silencing, ensuring spatiotemporal consistency of carrier and drug when shuttling multilayer tumor cells. As a result, the nanosponge achieves efficient tumor inhibition in 99.1% of tumor spheroids and 80.1% of orthotopic tumor models. Collectively, this study provides an intelligent nanosponge design for active intercellular relay drug delivery, achieving improved intratumoral bioavailability of drugs and amplified chemotherapy on solid tumors.
Collapse
Affiliation(s)
- Xiaoyu Yang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chenshuang Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Mingjie Song
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | | | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Huaqing Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| |
Collapse
|
39
|
Zhou Y, Chang W, Lu X, Wang J, Zhang C, Xu Y. Acid-base Homeostasis and Implications to the Phenotypic Behaviors of Cancer. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1133-1148. [PMID: 35787947 PMCID: PMC11082410 DOI: 10.1016/j.gpb.2022.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 05/27/2022] [Accepted: 06/26/2022] [Indexed: 12/23/2022]
Abstract
Acid-base homeostasis is a fundamental property of living cells, and its persistent disruption in human cells can lead to a wide range of diseases. In this study, we conducted a computational modeling analysis of transcriptomic data of 4750 human tissue samples of 9 cancer types in The Cancer Genome Atlas (TCGA) database. Built on our previous study, we quantitatively estimated the average production rate of OH- by cytosolic Fenton reactions, which continuously disrupt the intracellular pH (pHi) homeostasis. Our predictions indicate that all or at least a subset of 43 reprogrammed metabolisms (RMs) are induced to produce net protons (H+) at comparable rates of Fenton reactions to keep the pHi stable. We then discovered that a number of well-known phenotypes of cancers, including increased growth rate, metastasis rate, and local immune cell composition, can be naturally explained in terms of the Fenton reaction level and the induced RMs. This study strongly suggests the possibility to have a unified framework for studies of cancer-inducing stressors, adaptive metabolic reprogramming, and cancerous behaviors. In addition, strong evidence is provided to demonstrate that a popular view that Na+/H+ exchangers along with lactic acid exporters and carbonic anhydrases are responsible for the intracellular alkalization and extracellular acidification in cancer may not be justified.
Collapse
Affiliation(s)
- Yi Zhou
- Cancer Systems Biology Center, China-Japan Union Hospital, Jilin University, Changchun 130033, China; Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Wennan Chang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Xiaoyu Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biohealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Jin Wang
- Departments of Chemistry and of Physics and Astronomy, Stony Brook University, Stony Brook, NY 11794, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Ying Xu
- Cancer Systems Biology Center, China-Japan Union Hospital, Jilin University, Changchun 130033, China; Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
40
|
Tan R, Zhou Y, An Z, Xu Y. Cancer Is A Survival Process under Persistent Microenvironmental and Cellular Stresses. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1260-1265. [PMID: 35728722 PMCID: PMC11082257 DOI: 10.1016/j.gpb.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Renbo Tan
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Changchun 130000, China; College of Computer Science and Technology, Jilin University, Changchun 130000, China
| | - Yi Zhou
- Department of Biochemistry and Molecular Biology, and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Zheng An
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Changchun 130000, China; Department of Biochemistry and Molecular Biology, and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Ying Xu
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Changchun 130000, China; Department of Biochemistry and Molecular Biology, and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
41
|
Pazin WM, Miranda RR, Toledo KA, Kjeldsen F, Constantino CJL, Brewer JR. pH-Dependence Cytotoxicity Evaluation of Artepillin C against Tumor Cells. Life (Basel) 2023; 13:2186. [PMID: 38004326 PMCID: PMC10672498 DOI: 10.3390/life13112186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Brazilian green propolis is a well-known product that is consumed globally. Its major component, Artepillin C, showed potential as an antitumor product. This study explored the impact of Artepillin C on fibroblast and glioblastoma cell lines, used as healthy and very aggressive tumor cell lines, respectively. The focus of the study was to evaluate the pH-dependence of Artepillin C cytotoxicity, since tumor cells are known to have a more acidic extracellular microenvironment compared to healthy cells, and Artepillin C was shown to become more lipophilic at lower pH values. Investigations into the pH-dependency of Artepillin C (6.0-7.4), through viability assays and live cell imaging, revealed compelling insights. At pH 6.0, MTT assays showed the pronounced cytotoxic effects of Artepillin C, yielding a notable reduction in cell viability to less than 12% among glioblastoma cells following a 24 h exposure to 100 µM of Artepillin C. Concurrently, LDH assays indicated significant membrane damage, affecting approximately 50% of the total cells under the same conditions. Our Laurdan GP analysis suggests that Artepillin C induces autophagy, and notably, provokes a lipid membrane packing effect, contributing to cell death. These combined results affirm the selective cytotoxicity of Artepillin C within the acidic tumor microenvironment, emphasizing its potential as an effective antitumor agent. Furthermore, our findings suggest that Artepillin C holds promise for potential applications in the realm of anticancer therapies given its pH-dependence cytotoxicity.
Collapse
Affiliation(s)
- Wallance M. Pazin
- Department of Physics and Meteorology, School of Sciences, São Paulo State University (UNESP), Bauru 17033-360, Brazil;
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (R.R.M.); (F.K.)
| | - Renata R. Miranda
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (R.R.M.); (F.K.)
| | - Karina A. Toledo
- Department of Biological Sciences, School of Sciences, Humanities and Languages, São Paulo State University (UNESP), Assis 19806-900, Brazil;
| | - Frank Kjeldsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (R.R.M.); (F.K.)
| | - Carlos J. L. Constantino
- Department of Physics, School of Sciences and Technology, São Paulo State University (UNESP), Presidente Prudente 19060-900, Brazil;
| | - Jonathan R. Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (R.R.M.); (F.K.)
| |
Collapse
|
42
|
Guy L, Mosser M, Pitrat D, Mulatier JC, Kukułka M, Srebro-Hooper M, Jeanneau E, Bensalah-Ledoux A, Baguenard B, Guy S. Acid/Base-Triggered Photophysical and Chiroptical Switching in a Series of Helicenoid Compounds. Molecules 2023; 28:7322. [PMID: 37959742 PMCID: PMC10647711 DOI: 10.3390/molecules28217322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
A series of molecules that possess two quinolines, benzoquinolines, or phenanthrolines connected in a chiral fashion by a biaryl junction along with their water-soluble derivatives was developed and characterized. The influence of the structure on the basicity of the nitrogen atoms in two heterocycles was examined and the photophysical and chiroptical switching activity of the compounds upon protonation was studied both experimentally and computationally. The results demonstrated that changes in the electronic structure of the protonated vs. neutral species, promoting a bathochromic shift of dominant electronic transitions and alternation of their character from π-to-π* to charge-transfer-type, when additionally accompanied by the high structural flexibility of a system, leading to changes in conformational preferences upon proton binding, produce particularly pronounced modifications of the spectral properties in acidic medium. The latter combined with reversibility of the read-out make some of the molecules in this series very promising multifunctional pH probes.
Collapse
Affiliation(s)
- Laure Guy
- Laboratoire de Chimie UMR 5182, Université Lyon, ENS de Lyon, CNRS, F-69342 Lyon, France; (M.M.); (D.P.); (J.-C.M.)
| | - Maëlle Mosser
- Laboratoire de Chimie UMR 5182, Université Lyon, ENS de Lyon, CNRS, F-69342 Lyon, France; (M.M.); (D.P.); (J.-C.M.)
| | - Delphine Pitrat
- Laboratoire de Chimie UMR 5182, Université Lyon, ENS de Lyon, CNRS, F-69342 Lyon, France; (M.M.); (D.P.); (J.-C.M.)
| | - Jean-Christophe Mulatier
- Laboratoire de Chimie UMR 5182, Université Lyon, ENS de Lyon, CNRS, F-69342 Lyon, France; (M.M.); (D.P.); (J.-C.M.)
| | - Mercedes Kukułka
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland
| | | | - Erwann Jeanneau
- Centre de Diffractométrie Henri Longchambon, Université Claude Bernard Lyon 1, 5 Rue de la Doua, F-69100 Villeurbanne, France;
| | - Amina Bensalah-Ledoux
- Institut Lumière Matière UMR 5306, Université Lyon, CNRS, F-69622 Villeurbanne, France; (A.B.-L.); (B.B.); (S.G.)
| | - Bruno Baguenard
- Institut Lumière Matière UMR 5306, Université Lyon, CNRS, F-69622 Villeurbanne, France; (A.B.-L.); (B.B.); (S.G.)
| | - Stéphan Guy
- Institut Lumière Matière UMR 5306, Université Lyon, CNRS, F-69622 Villeurbanne, France; (A.B.-L.); (B.B.); (S.G.)
| |
Collapse
|
43
|
Henoumont C, Devreux M, Laurent S. Mn-Based MRI Contrast Agents: An Overview. Molecules 2023; 28:7275. [PMID: 37959694 PMCID: PMC10648041 DOI: 10.3390/molecules28217275] [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: 09/29/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
MRI contrast agents are required in the clinic to detect some pathologies, such as cancers. Nevertheless, at the moment, only small extracellular and non-specific gadolinium complexes are available for clinicians. Moreover, safety issues have recently emerged concerning the use of gadolinium complexes; hence, alternatives are urgently needed. Manganese-based MRI contrast agents could be one of these alternatives and increasing numbers of studies are available in the literature. This review aims at synthesizing all the research, from small Mn complexes to nanoparticular agents, including theranostic agents, to highlight all the efforts already made by the scientific community to obtain highly efficient agents but also evidence of the weaknesses of the developed systems.
Collapse
Affiliation(s)
- Céline Henoumont
- NMR and Molecular Imaging Laboratory, Department of General, Organic and Biomedical Chemistry, University of Mons, 19 Avenue Maistriau, 7000 Mons, Belgium; (C.H.)
| | - Marie Devreux
- NMR and Molecular Imaging Laboratory, Department of General, Organic and Biomedical Chemistry, University of Mons, 19 Avenue Maistriau, 7000 Mons, Belgium; (C.H.)
| | - Sophie Laurent
- NMR and Molecular Imaging Laboratory, Department of General, Organic and Biomedical Chemistry, University of Mons, 19 Avenue Maistriau, 7000 Mons, Belgium; (C.H.)
- Center for Microscopy and Molecular Imaging (CMMI), 8 Rue Adrienne Boland, 6041 Gosselies, Belgium
| |
Collapse
|
44
|
Li H, Peng T, Zhong Y, Liu M, Mak PI, Martins RP, Wang P, Jia Y. pH Regulator on Digital Microfluidics with Pico-Dosing Technique. BIOSENSORS 2023; 13:951. [PMID: 37998126 PMCID: PMC10669492 DOI: 10.3390/bios13110951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 11/25/2023]
Abstract
Real-time pH control on-chip is a crucial factor for cell-based experiments in microfluidics, yet difficult to realize. In this paper, we present a flexible pH regulator on a digital microfluidic (DMF) platform. The pico-dosing technology, which can generate and transfer satellite droplets, is presented to deliver alkali/acid into the sample solution to change the pH value of the sample. An image analysis method based on ImageJ is developed to calculate the delivered volume and an on-chip colorimetric method is proposed to determine the pH value of the sample solution containing the acid-base indicator. The calculated pH values show consistency with the measured ones. Our approach makes the real-time pH control of the on-chip biological experiment more easy to control and flexible.
Collapse
Affiliation(s)
- Haoran Li
- The State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau 999078, China; (H.L.); (M.L.); (P.-I.M.)
| | - Tao Peng
- Zhuhai UM Science & Technology Research Institute, Zhuhai 519085, China;
| | - Yunlong Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Meiqing Liu
- The State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau 999078, China; (H.L.); (M.L.); (P.-I.M.)
| | - Pui-In Mak
- The State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau 999078, China; (H.L.); (M.L.); (P.-I.M.)
- Zhuhai UM Science & Technology Research Institute, Zhuhai 519085, China;
| | - Rui P. Martins
- The State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau 999078, China; (H.L.); (M.L.); (P.-I.M.)
- Faculty of Science and Technology–ECE, University of Macau, Macau 999078, China
- Instituto Superior Técnico, Unversidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Ping Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yanwei Jia
- The State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau 999078, China; (H.L.); (M.L.); (P.-I.M.)
- Faculty of Science and Technology–ECE, University of Macau, Macau 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| |
Collapse
|
45
|
Alonso-Carrillo D, Arias-Betancur A, Carreira-Barral I, Fontova P, Soto-Cerrato V, García-Valverde M, Pérez-Tomás R, Quesada R. Small molecule anion carriers facilitate lactate transport in model liposomes and cells. iScience 2023; 26:107898. [PMID: 37790273 PMCID: PMC10543179 DOI: 10.1016/j.isci.2023.107898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 10/05/2023] Open
Abstract
An excessive production of lactate by cancer cells fosters tumor growth and metastasis. Therefore, targeting lactate metabolism and transport offers a new therapeutic strategy against cancer, based on dependency of some cancer cells for lactate as energy fuel or as oncogenic signal. Herein we present a family of anionophores based on the structure of click-tambjamines that have proved to be extremely active lactate carriers across phospholipid membranes. Compound 1, the most potent lactate transmembrane carrier, was studied in HeLa cells. The use of a monocarboxylate transporters (MCTs) inhibitor proved that 1 is an active lactate transporter in living cells, confirming the results obtained in phospholipid vesicles. Moreover, an additive effect of compound 1 with cisplatin was observed in HeLa cells. Identification of active lactate anionophores working in living cells opens up ways to exploit this class of compounds as molecular tools and drugs addressing dysregulated lactate metabolism.
Collapse
Affiliation(s)
- Daniel Alonso-Carrillo
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Alain Arias-Betancur
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, L’Hospitalet de Llobregat, 08907, Spain
- Molecular Signalling, Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908, Spain
- Department of Integral Adult Dentistry, Dental School, Research Centre for Dental Sciences, Universidad de La Frontera, Temuco 4811230, Chile
| | - Israel Carreira-Barral
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Pere Fontova
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, L’Hospitalet de Llobregat, 08907, Spain
- Molecular Signalling, Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908, Spain
| | - María García-Valverde
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, L’Hospitalet de Llobregat, 08907, Spain
- Molecular Signalling, Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908, Spain
| | - Roberto Quesada
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| |
Collapse
|
46
|
Ahmed Z, LoGiudice K, Mays G, Schorr A, Rowey R, Yang H, Trivedi S, Srivastava V. Increasing Chemotherapeutic Efficacy Using pH-Modulating and Doxorubicin-Releasing Injectable Chitosan-Poly(ethylene glycol) Hydrogels. ACS APPLIED MATERIALS & INTERFACES 2023; 15:45626-45639. [PMID: 37729014 DOI: 10.1021/acsami.3c09733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Modulation of pH is crucial to maintaining the chemical homeostasis of biological environments. The irregular metabolic pathways exhibited by cancer cells result in the production of acidic byproducts that are excreted and accumulate in the extracellular tumor microenvironment, reducing the pH. As a consequence of the lower pH in tumors, cancer cells increase the expression of metastatic phenotypes and chemotherapeutic resistance. A significant limitation in current cancer therapies is the inability to locally deliver chemotherapeutics, leading to significant damage to healthy cells in systemic administration. To overcome these challenges, we present an injectable chitosan-poly(ethylene glycol) hydrogel that is dual-loaded with doxorubicin and sodium bicarbonate providing alkaline buffering of extracellular acidity and simultaneous chemotherapeutic delivery to increase chemotherapeutic efficacy. We conducted in vitro studies of weak base chemotherapeutic and alkaline buffer release from the hydrogel. The release of doxorubicin from hydrogels increased in a low-pH environment and was dependent on the encapsulated sodium bicarbonate concentration. We investigated the influence of pH on the doxorubicin efficacy and viability of MCF-7 and MDA-MB-231 breast cancer cell lines. The results show a 2- to 3-fold increase in IC50 values from neutral pH to low pH, showing decreased cancer cell viability at neutral pH as compared to acidic pH. The IC50 results were shown to correlate with a decrease in intracellular uptake of doxorubicin at low pH. The proposed hydrogels were confirmed to be nontoxic to healthy MCF-10A mammary epithelial cells. Rheological studies were performed to verify that the dual-loaded hydrogels were injectable. The mechanical and release properties of the hydrogels were maintained after extended storage. The chemotherapeutic activity of doxorubicin was evaluated in the presence of the proposed pH-regulating hydrogels. The findings suggest a promising nontoxic, biodegradable hydrogel buffer delivery system that can achieve two simultaneous important goals of local acidosis neutralization and chemotherapeutic release.
Collapse
Affiliation(s)
- Zahra Ahmed
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Kevin LoGiudice
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Gavin Mays
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Angelina Schorr
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Rachel Rowey
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Haisong Yang
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Shruti Trivedi
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Vikas Srivastava
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
47
|
Malcolm JR, Sajjaboontawee N, Yerlikaya S, Plunkett-Jones C, Boxall PJ, Brackenbury WJ. Voltage-gated sodium channels, sodium transport and progression of solid tumours. CURRENT TOPICS IN MEMBRANES 2023; 92:71-98. [PMID: 38007270 DOI: 10.1016/bs.ctm.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Sodium (Na+) concentration in solid tumours of different origin is highly dysregulated, and this corresponds to the aberrant expression of Na+ transporters. In particular, the α subunits of voltage gated Na+ channels (VGSCs) raise intracellular Na+ concentration ([Na+]i) in malignant cells, which influences the progression of solid tumours, predominantly driving cancer cells towards a more aggressive and metastatic phenotype. Conversely, re-expression of VGSC β subunits in cancer cells can either enhance tumour progression or promote anti-tumourigenic properties. Metastasis is the leading cause of cancer-related mortality, highlighting an important area of research which urgently requires improved therapeutic interventions. Here, we review the extent to which VGSC subunits are dysregulated in solid tumours, and consider the implications of such dysregulation on solid tumour progression. We discuss current understanding of VGSC-dependent mechanisms underlying increased invasive and metastatic potential of solid tumours, and how the complex relationship between the tumour microenvironment (TME) and VGSC expression may further drive tumour progression, in part due to the interplay of infiltrating immune cells, cancer-associated fibroblasts (CAFs) and insufficient supply of oxygen (hypoxia). Finally, we explore past and present clinical trials that investigate utilising existing VGSC modulators as potential pharmacological options to support adjuvant chemotherapies to prevent cancer recurrence. Such research demonstrates an exciting opportunity to repurpose therapeutics in order to improve the disease-free survival of patients with aggressive solid tumours.
Collapse
Affiliation(s)
- Jodie R Malcolm
- Department of Biology, University of York, Heslington, York, United Kingdom
| | - Nattanan Sajjaboontawee
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom
| | - Serife Yerlikaya
- Department of Biology, University of York, Heslington, York, United Kingdom; Istanbul Medipol University, Research Institute for Health Sciences and Technologies, Istanbul, Turkey
| | | | - Peter J Boxall
- Department of Biology, University of York, Heslington, York, United Kingdom; York and Scarborough Teaching Hospitals NHS Foundation Trust, York, United Kingdom
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom.
| |
Collapse
|
48
|
Bastos FR, Soares da Costa D, Reis RL, Alves NM, Pashkuleva I, Costa RR. Layer-by-layer coated calcium carbonate nanoparticles for targeting breast cancer cells. BIOMATERIALS ADVANCES 2023; 153:213563. [PMID: 37487456 DOI: 10.1016/j.bioadv.2023.213563] [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: 05/09/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Breast cancer is resistant to conventional treatments due to the specific tumour microenvironment, the associated acidic pH and the overexpression of receptors that enhance cells tumorigenicity. Herein, we optimized the synthesis of acidic resorbable calcium carbonate (CaCO3) nanoparticles and the encapsulation of a low molecular weight model molecule (Rhodamine). The addition of ethylene glycol during the synthetic process resulted in a particle size decrease: we obtained homogeneous CaCO3 particles with an average size of 564 nm. Their negative charge enabled the assembly of layer-by-layer (LbL) coatings with surface-exposed hyaluronic acid (HA), a ligand of tumour-associated receptor CD44. The coating decreased Rhodamine release by two-fold compared to uncoated nanoparticles. We demonstrated the effect of nanoparticles on two breast cancer cell lines with different aggressiveness - SK-BR-3 and the more aggressive MDA-MB-231 - and compared them with the normal breast cell line MCF10A. CaCO3 nanoparticles (coated and uncoated) significantly decreased the metabolic activity of the breast cancer cells. The interactions between LbL-coated nanoparticles and cells depended on HA expression on the cell surface: more particles were observed on the surface of MDA-MB-231 cells, which had the thickest endogenous HA coating. We concluded that CaCO3 nanoparticles are potential candidates to carry low molecular weight chemotherapeutics and deliver them to aggressive breast cancer sites with an HA-abundant pericellular matrix.
Collapse
Affiliation(s)
- Filipa R Bastos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Natália M Alves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Rui R Costa
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
49
|
Cornwell AC, Tisdale AA, Venkat S, Maraszek KE, Alahmari AA, George A, Attwood K, George M, Rempinski D, Franco-Barraza J, Seshadri M, Parker MD, Cortes Gomez E, Fountzilas C, Cukierman E, Steele NG, Feigin ME. Lorazepam Stimulates IL6 Production and Is Associated with Poor Survival Outcomes in Pancreatic Cancer. Clin Cancer Res 2023; 29:3793-3812. [PMID: 37587561 PMCID: PMC10502465 DOI: 10.1158/1078-0432.ccr-23-0547] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/31/2023] [Accepted: 07/19/2023] [Indexed: 08/18/2023]
Abstract
PURPOSE This research investigates the association between benzodiazepines (BZD) and cancer patient survival outcomes, the pancreatic cancer tumor microenvironment, and cancer-associated fibroblast (CAF) signaling. EXPERIMENTAL DESIGN Multivariate Cox regression modeling was used to retrospectively measure associations between Roswell Park cancer patient survival outcomes and BZD prescription records. IHC, H&E, Masson's trichrome, RNAscope, and RNA sequencing were used to evaluate the impact of lorazepam (LOR) on the murine PDAC tumor microenvironment. ELISA and qPCR were used to determine the impact of BZDs on IL6 expression or secretion by human-immortalized pancreatic CAFs. PRESTO-Tango assays, reanalysis of PDAC single-cell sequencing/TCGA data sets, and GPR68 CRISPRi knockdown CAFs were used to determine the impact of BZDs on GPR68 signaling. RESULTS LOR is associated with worse progression-free survival (PFS), whereas alprazolam (ALP) is associated with improved PFS, in pancreatic cancer patients receiving chemotherapy. LOR promotes desmoplasia (fibrosis and extracellular matrix protein deposition), inflammatory signaling, and ischemic necrosis. GPR68 is preferentially expressed on human PDAC CAFs, and n-unsubstituted BZDs, such as LOR, significantly increase IL6 expression and secretion in CAFs in a pH and GPR68-dependent manner. Conversely, ALP and other GPR68 n-substituted BZDs decrease IL6 in human CAFs in a pH and GPR68-independent manner. Across many cancer types, LOR is associated with worse survival outcomes relative to ALP and patients not receiving BZDs. CONCLUSIONS We demonstrate that LOR stimulates fibrosis and inflammatory signaling, promotes desmoplasia and ischemic necrosis, and is associated with decreased pancreatic cancer patient survival.
Collapse
Affiliation(s)
- Abigail C. Cornwell
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Arwen A. Tisdale
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Swati Venkat
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Kathryn E. Maraszek
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Abdulrahman A. Alahmari
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Anthony George
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Madison George
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Donald Rempinski
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Janusz Franco-Barraza
- Cancer Signaling and Microenvironment Program, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Marvin and Concetta Greenberg Pancreatic Cancer Institute, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mukund Seshadri
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Mark D. Parker
- Department of Physiology and Biophysics, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
- Department of Ophthalmology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Marvin and Concetta Greenberg Pancreatic Cancer Institute, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Nina G. Steele
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Michael E. Feigin
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
50
|
Munan S, Yadav R, Pareek N, Samanta A. Ratiometric fluorescent probes for pH mapping in cellular organelles. Analyst 2023; 148:4242-4262. [PMID: 37581493 DOI: 10.1039/d3an00960b] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
The intracellular pH (pHi) in organelles, including mitochondria, endoplasmic reticulum, lysosomes, and nuclei, differs from the cytoplasmic pH, and thus maintaining the pH of these organelles is crucial for cellular homeostasis. Alterations in the intracellular pH (ΔpHi) in organelles lead to the disruption of cell proliferation, ion transportation, cellular homeostasis, and even cell death. Hence, accurately mapping the pH of organelles is crucial. Accordingly, the development of fluorescence imaging probes for targeting specific organelles and monitoring their dynamics at the molecular level has become the forefront of research in the last three decades. Among them, ratiometric fluorescent probes minimize the interference from the excitation wavelength of light, auto-fluorescence from probe concentration, environmental fluctuations, and instrument sensitivity through self-correction compared to monochromatic fluorescent probes, which are known as turn-on/off fluorescent probes. Small-molecular ratiometric fluorescent probes for detecting ΔpHi are challenging yet demanding. To date, sixty-two ratiometric pH probes have been reported for monitoring internal pH alterations in cellular organelles. However, a critical review on organelle-specific ratiometric probes for pH mapping is still lacking. Thus, in the present review, we report the most recent advances in ratiometric pH probes and the previous data on the role of mapping the ΔpHi of cellular organelles. The development strategy, including ratiometric fluorescence with one reference signal (RFRS) and ratiometric fluorescence with two reversible signals (RFRvS), is systematically illustrated. Finally, we emphasize the major challenges in developing ratiometric probes that merit further research in the future.
Collapse
Affiliation(s)
- Subrata Munan
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institute of Eminence Deemed to be University, NH 91, Tehsil Dadri, Uttar Pradesh, India 201314.
| | - Rashmi Yadav
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institute of Eminence Deemed to be University, NH 91, Tehsil Dadri, Uttar Pradesh, India 201314.
| | - Niharika Pareek
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institute of Eminence Deemed to be University, NH 91, Tehsil Dadri, Uttar Pradesh, India 201314.
| | - Animesh Samanta
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institute of Eminence Deemed to be University, NH 91, Tehsil Dadri, Uttar Pradesh, India 201314.
| |
Collapse
|