1
|
Kamarulzaman NT, Makpol S. The link between Mitochondria and Sarcopenia. J Physiol Biochem 2025; 81:1-20. [PMID: 39969761 PMCID: PMC11958477 DOI: 10.1007/s13105-024-01062-7] [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: 05/28/2024] [Accepted: 11/12/2024] [Indexed: 02/20/2025]
Abstract
Sarcopenia, a widespread condition, is characterized by a variety of factors influencing its development. The causes of sarcopenia differ depending on the age of the individual. It is defined as the combination of decreased muscle mass and impaired muscle function, primarily observed in association with ageing. As people age from 20 to 80 years old, there is an approximate 30% reduction in muscle mass and a 20% decline in cross-sectional area. This decline is attributed to a decrease in the size and number of muscle fibres. The regression of muscle mass and strength increases the risk of fractures, frailty, reduced quality of life, and loss of independence. Muscle cells, fibres, and tissues shrink, resulting in diminished muscle power, volume, and strength in major muscle groups. One prominent theory of cellular ageing posits a strong positive relationship between age and oxidative damage. Heightened oxidative stress leads to early-onset sarcopenia, characterized by neuromuscular innervation breakdown, muscle atrophy, and dysfunctional mitochondrial muscles. Ageing muscles generate more reactive oxygen species (ROS), and experience decreased oxygen consumption and ATP synthesis compared to younger muscles. Additionally, changes in mitochondrial protein interactions, cristae structure, and networks may contribute to ADP insensitivity, which ultimately leads to sarcopenia. Within this framework, this review provides a comprehensive summary of our current understanding of the role of mitochondria in sarcopenia and other muscle degenerative diseases, highlighting the crucial need for further research in these areas.
Collapse
Affiliation(s)
- Nurul Tihani Kamarulzaman
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia.
| |
Collapse
|
2
|
Komza M, Chipuk JE. Mitochondrial metabolism: A moving target in hepatocellular carcinoma therapy. J Cell Physiol 2025; 240:e31441. [PMID: 39324415 PMCID: PMC11732733 DOI: 10.1002/jcp.31441] [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: 05/01/2024] [Revised: 08/21/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Mitochondria are pivotal contributors to cancer mechanisms due to their homeostatic and pathological roles in cellular bioenergetics, biosynthesis, metabolism, signaling, and survival. During transformation and tumor initiation, mitochondrial function is often disrupted by oncogenic mutations, leading to a metabolic profile distinct from precursor cells. In this review, we focus on hepatocellular carcinoma, a cancer arising from metabolically robust and nutrient rich hepatocytes, and discuss the mechanistic impact of altered metabolism in this setting. We provide distinctions between normal mitochondrial activity versus disease-related function which yielded therapeutic opportunities, along with highlighting recent preclinical and clinical efforts focused on targeting mitochondrial metabolism. Finally, several novel strategies for exploiting mitochondrial programs to eliminate hepatocellular carcinoma cells in metabolism-specific contexts are presented to integrate these concepts and gain foresight into the future of mitochondria-focused therapeutics.
Collapse
Affiliation(s)
- Monika Komza
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, New York, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn School of Medicine at Mount Sinai, The Diabetes, Obesity, and Metabolism Institute, New York, New York, USA
| |
Collapse
|
3
|
Comolet G, Bose N, Winchell J, Duren-Lubanski A, Rusielewicz T, Goldberg J, Horn G, Paull D, Migliori B. A highly efficient, scalable pipeline for fixed feature extraction from large-scale high-content imaging screens. iScience 2024; 27:111434. [PMID: 39720532 PMCID: PMC11667173 DOI: 10.1016/j.isci.2024.111434] [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: 10/02/2023] [Revised: 08/15/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Applying artificial intelligence (AI) to image-based morphological profiling cells offers significant potential for identifying disease states and drug responses in high-content imaging (HCI) screens. When differences between populations (e.g., healthy vs. diseased) are unknown or imperceptible to the human eye, large-scale HCI screens are essential, providing numerous replicates to build reliable models and accounting for confounding factors like donor and intra-experimental variations. As screen sizes grow, so does the challenge of analyzing high-dimensional datasets in an efficient way while preserving interpretable features and predictive power. Here, we introduce ScaleFEx℠, a memory-efficient, open-source Python pipeline that extracts biologically meaningful features from HCI datasets using minimal computational resources or scalable cloud infrastructure. ScaleFEx can be used together with AI models to successfully identify phenotypic shifts in drug-treated cells and rank interpretable features, and is applicable to public datasets, highlighting its potential to accelerate the discovery of disease-associated phenotypes and new therapeutics.
Collapse
Affiliation(s)
- Gabriel Comolet
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Neeloy Bose
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Jeff Winchell
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | | | - Tom Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Jordan Goldberg
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Grayson Horn
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Bianca Migliori
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| |
Collapse
|
4
|
Handley S, Anwer AG, Knab A, Bhargava A, Goldys EM. AutoMitoNetwork: Software for analyzing mitochondrial networks in autofluorescence images to enable label-free cell classification. Cytometry A 2024; 105:688-703. [PMID: 39078083 DOI: 10.1002/cyto.a.24889] [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: 03/04/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024]
Abstract
High-resolution mitochondria imaging in combination with image analysis tools have significantly advanced our understanding of cellular function in health and disease. However, most image analysis tools for mitochondrial studies have been designed to work with fluorescently labeled images only. Additionally, efforts to integrate features describing mitochondrial networks with machine learning techniques for the differentiation of cell types have been limited. Herein, we present AutoMitoNetwork software for image-based assessment of mitochondrial networks in label-free autofluorescence images using a range of interpretable morphological, intensity, and textural features. To demonstrate its utility, we characterized unstained mitochondrial networks in healthy retinal cells and in retinal cells exposed to two types of treatments: rotenone, which directly inhibited mitochondrial respiration and ATP production, and iodoacetic acid, which had a milder impact on mitochondrial networks via the inhibition of anaerobic glycolysis. For both cases, our multi-dimensional feature analysis combined with a support vector machine classifier distinguished between healthy cells and those treated with rotenone or iodoacetic acid. Subtle changes in morphological features were measured including increased fragmentation in the treated retinal cells, pointing to an association with metabolic mechanisms. AutoMitoNetwork opens new options for image-based machine learning in label-free imaging, diagnostics, and mitochondrial disease drug development.
Collapse
Affiliation(s)
- Shannon Handley
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), University of New South Wales, Sydney, New South Wales, Australia
- The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | - Ayad G Anwer
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), University of New South Wales, Sydney, New South Wales, Australia
- The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | - Aline Knab
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), University of New South Wales, Sydney, New South Wales, Australia
- The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | - Akanksha Bhargava
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), University of New South Wales, Sydney, New South Wales, Australia
- The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | - Ewa M Goldys
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), University of New South Wales, Sydney, New South Wales, Australia
- The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Yuan L, Cao Y, Zhang Q, Pan J, Wu C, Ye Y, Jiao Q, Zhu HL, Wang Z. Rational design of mitochondria-targeted fluorescent biosensors for in vivo elucidation of the interaction between breast cancer metastasis and mitochondrial autophagy. Biosens Bioelectron 2024; 251:116123. [PMID: 38359670 DOI: 10.1016/j.bios.2024.116123] [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: 12/21/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Breast cancer lung metastases (BCLM) are a major cause of high mortality in patients. The shortage of therapeutic targets and rapid drug screening tools for BCLM is a major challenge at present. Mitochondrial autophagy, which involves the degradation of proteins associated with cancer cell aggressiveness, represents a possible therapeutic approach for the treatment of BCLM. Herein, four fluorescent biosensors with different alkyl chains were designed and synthesized to monitor mitochondrial autophagy. Among them, PMV-12 demonstrated the highest sensitivity to viscosity variance, the least impact on polarity, and the longest imaging time. The introduction of the C12-chain made PMV-12 anchored in the mitochondrial membrane without being disturbed by changes of the mitochondrial membrane potential (MMP), thereby achieving the long-term monitor in situ for mitochondrial autophagy. Mitochondria stained with PMV-12 induced swelling and viscosity increase after treating with apigenin, which indicated that apigenin is a potential mitochondrial autophagy inducer. Apigenin was subsequently verified to inhibit cancer cell invasion by 92%. Furthermore, PMV-12 could monitor the process of BCLM in vivo and evaluate the therapeutic effects of apigenin. This work provides a fluorescent tool for elucidating the role of mitochondrial autophagy in the BCLM process and for anti-metastatic drug development.
Collapse
Affiliation(s)
- Liangchao Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China
| | - Yuyao Cao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China
| | - Qing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China
| | - Jiancheng Pan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China
| | - Changjian Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China
| | - Yaxi Ye
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, 234000, PR China.
| | - Qingcai Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China.
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China.
| | - Zhongchang Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, 210023, PR China.
| |
Collapse
|
6
|
Ichegiri A, Kodolikar K, Bagade V, Selukar M, Dey T. Mitochondria: A source of potential biomarkers for non-communicable diseases. Adv Clin Chem 2024; 121:334-365. [PMID: 38797544 DOI: 10.1016/bs.acc.2024.04.007] [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: 05/29/2024]
Abstract
Mitochondria, as an endosymbiont of eukaryotic cells, controls multiple cellular activities, including respiration, reactive oxygen species production, fatty acid synthesis, and death. Though the majority of functional mitochondrial proteins are translated through a nucleus-controlled process, very few of them (∼10%) are translated within mitochondria through their own machinery. Germline and somatic mutations in mitochondrial and nuclear DNA significantly impact mitochondrial homeostasis and function. Such modifications disturbing mitochondrial biogenesis, metabolism, or mitophagy eventually resulted in cellular pathophysiology. In this chapter, we discussed the impact of mitochondria and its dysfunction on several non-communicable diseases like cancer, diabetes, neurodegenerative, and cardiovascular problems. Mitochondrial dysfunction and its outcome could be screened by currently available omics-based techniques, flow cytometry, and high-resolution imaging. Such characterization could be evaluated as potential biomarkers to assess the disease burden and prognosis.
Collapse
Affiliation(s)
- Amulya Ichegiri
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Kshitij Kodolikar
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Vaibhavi Bagade
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Mrunal Selukar
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Tuli Dey
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India.
| |
Collapse
|
7
|
Li Y, Guo Z, Gao X, Wang G. MMCL-CDR: enhancing cancer drug response prediction with multi-omics and morphology images contrastive representation learning. Bioinformatics 2023; 39:btad734. [PMID: 38070154 PMCID: PMC10756335 DOI: 10.1093/bioinformatics/btad734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/09/2023] [Indexed: 12/30/2023] Open
Abstract
MOTIVATION Cancer is a complex disease that results in a significant number of global fatalities. Treatment strategies can vary among patients, even if they have the same type of cancer. The application of precision medicine in cancer shows promise for treating different types of cancer, reducing healthcare expenses, and improving recovery rates. To achieve personalized cancer treatment, machine learning models have been developed to predict drug responses based on tumor and drug characteristics. However, current studies either focus on constructing homogeneous networks from single data source or heterogeneous networks from multiomics data. While multiomics data have shown potential in predicting drug responses in cancer cell lines, there is still a lack of research that effectively utilizes insights from different modalities. Furthermore, effectively utilizing the multimodal knowledge of cancer cell lines poses a challenge due to the heterogeneity inherent in these modalities. RESULTS To address these challenges, we introduce MMCL-CDR (Multimodal Contrastive Learning for Cancer Drug Responses), a multimodal approach for cancer drug response prediction that integrates copy number variation, gene expression, morphology images of cell lines, and chemical structure of drugs. The objective of MMCL-CDR is to align cancer cell lines across different data modalities by learning cell line representations from omic and image data, and combined with structural drug representations to enhance the prediction of cancer drug responses (CDR). We have carried out comprehensive experiments and show that our model significantly outperforms other state-of-the-art methods in CDR prediction. The experimental results also prove that the model can learn more accurate cell line representation by integrating multiomics and morphological data from cell lines, thereby improving the accuracy of CDR prediction. In addition, the ablation study and qualitative analysis also confirm the effectiveness of each part of our proposed model. Last but not least, MMCL-CDR opens up a new dimension for cancer drug response prediction through multimodal contrastive learning, pioneering a novel approach that integrates multiomics and multimodal drug and cell line modeling. AVAILABILITY AND IMPLEMENTATION MMCL-CDR is available at https://github.com/catly/MMCL-CDR.
Collapse
Affiliation(s)
- Yang Li
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150006, China
| | - Zihou Guo
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150006, China
| | - Xin Gao
- Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150006, China
| |
Collapse
|
8
|
Charrasse S, Poquillon T, Saint-Omer C, Pastore M, Bordignon B, Frye RE, Reynes C, Racine V, Aouacheria A. Quantitative assessment of mitochondrial morphology relevant for studies on cellular health and environmental toxicity. Comput Struct Biotechnol J 2023; 21:5609-5619. [PMID: 38047232 PMCID: PMC10690410 DOI: 10.1016/j.csbj.2023.11.015] [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] [Indexed: 12/05/2023] Open
Abstract
Mitochondria are essential organelles that play crucial roles in cellular energy metabolism, calcium signaling and apoptosis. Their importance in tissue homeostasis and stress responses, combined to their ability to transition between various structural and functional states, make them excellent organelles for monitoring cellular health. Quantitative assessment of mitochondrial morphology can therefore provide valuable insights into environmentally-induced cell damage. High-content screening (HCS) provides a powerful tool for analyzing organelles and cellular substructures. We developed a fully automated and miniaturized HCS wet-plus-dry pipeline (MITOMATICS) exploiting mitochondrial morphology as a marker for monitoring cellular health or damage. MITOMATICS uses an in-house, proprietary software (MitoRadar) to enable fast, exhaustive and cost-effective analysis of mitochondrial morphology and its inherent diversity in live cells. We applied our pipeline and big data analytics software to assess the mitotoxicity of selected chemicals, using the mitochondrial uncoupler CCCP as an internal control. Six different pesticides (inhibiting complexes I, II and III of the mitochondrial respiratory chain) were tested as individual compounds and five other pesticides present locally in Occitanie (Southern France) were assessed in combination to determine acute mitotoxicity. Our results show that the assayed pesticides exhibit specific signatures when used as single compounds or chemical mixtures and that they function synergistically to impact mitochondrial architecture. Study of environment-induced mitochondrial damage has the potential to open new fields in mechanistic toxicology, currently underexplored by regulatory toxicology and exposome research. Such exploration could inform health policy guidelines and foster pharmacological intervention, water, air and soil pollution control and food safety.
Collapse
Affiliation(s)
- Sophie Charrasse
- Institut des Sciences de l′Evolution de Montpellier (ISEM, UMR 5554, CNRS/UM/IRD/EPHE), Université de Montpellier, Montpellier, France
| | - Titouan Poquillon
- Institut des Sciences de l′Evolution de Montpellier (ISEM, UMR 5554, CNRS/UM/IRD/EPHE), Université de Montpellier, Montpellier, France
- QuantaCell SAS, Hôpital Saint Eloi, IRMB, 80 avenue Augustin Fliche, 34090 Montpellier, France
| | - Charlotte Saint-Omer
- Institut des Sciences de l′Evolution de Montpellier (ISEM, UMR 5554, CNRS/UM/IRD/EPHE), Université de Montpellier, Montpellier, France
| | - Manuela Pastore
- STATABIO BioCampus, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Benoit Bordignon
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Christelle Reynes
- STATABIO BioCampus, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Victor Racine
- QuantaCell SAS, Hôpital Saint Eloi, IRMB, 80 avenue Augustin Fliche, 34090 Montpellier, France
| | - Abdel Aouacheria
- Institut des Sciences de l′Evolution de Montpellier (ISEM, UMR 5554, CNRS/UM/IRD/EPHE), Université de Montpellier, Montpellier, France
| |
Collapse
|
9
|
Chen L, Zhou M, Li H, Liu D, Liao P, Zong Y, Zhang C, Zou W, Gao J. Mitochondrial heterogeneity in diseases. Signal Transduct Target Ther 2023; 8:311. [PMID: 37607925 PMCID: PMC10444818 DOI: 10.1038/s41392-023-01546-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 08/24/2023] Open
Abstract
As key organelles involved in cellular metabolism, mitochondria frequently undergo adaptive changes in morphology, components and functions in response to various environmental stresses and cellular demands. Previous studies of mitochondria research have gradually evolved, from focusing on morphological change analysis to systematic multiomics, thereby revealing the mitochondrial variation between cells or within the mitochondrial population within a single cell. The phenomenon of mitochondrial variation features is defined as mitochondrial heterogeneity. Moreover, mitochondrial heterogeneity has been reported to influence a variety of physiological processes, including tissue homeostasis, tissue repair, immunoregulation, and tumor progression. Here, we comprehensively review the mitochondrial heterogeneity in different tissues under pathological states, involving variant features of mitochondrial DNA, RNA, protein and lipid components. Then, the mechanisms that contribute to mitochondrial heterogeneity are also summarized, such as the mutation of the mitochondrial genome and the import of mitochondrial proteins that result in the heterogeneity of mitochondrial DNA and protein components. Additionally, multiple perspectives are investigated to better comprehend the mysteries of mitochondrial heterogeneity between cells. Finally, we summarize the prospective mitochondrial heterogeneity-targeting therapies in terms of alleviating mitochondrial oxidative damage, reducing mitochondrial carbon stress and enhancing mitochondrial biogenesis to relieve various pathological conditions. The possibility of recent technological advances in targeted mitochondrial gene editing is also discussed.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengnan Zhou
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, 110001, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China.
| |
Collapse
|
10
|
Wang L, Goldwag J, Bouyea M, Barra J, Matteson K, Maharjan N, Eladdadi A, Embrechts MJ, Intes X, Kruger U, Barroso M. Spatial topology of organelle is a new breast cancer cell classifier. iScience 2023; 26:107229. [PMID: 37519903 PMCID: PMC10384275 DOI: 10.1016/j.isci.2023.107229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 05/10/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Genomics and proteomics have been central to identify tumor cell populations, but more accurate approaches to classify cell subtypes are still lacking. We propose a new methodology to accurately classify cancer cells based on their organelle spatial topology. Herein, we developed an organelle topology-based cell classification pipeline (OTCCP), which integrates artificial intelligence (AI) and imaging quantification to analyze organelle spatial distribution and inter-organelle topology. OTCCP was used to classify a panel of human breast cancer cells, grown as 2D monolayer or 3D tumor spheroids using early endosomes, mitochondria, and their inter-organelle contacts. Organelle topology allows for a highly precise differentiation between cell lines of different subtypes and aggressiveness. These findings lay the groundwork for using organelle topological profiling as a fast and efficient method for phenotyping breast cancer function as well as a discovery tool to advance our understanding of cancer cell biology at the subcellular level.
Collapse
Affiliation(s)
- Ling Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Joshua Goldwag
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Megan Bouyea
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Jonathan Barra
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Kailie Matteson
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Niva Maharjan
- Department of Mathematics, The College of Saint Rose, Albany, NY 12203, USA
| | - Amina Eladdadi
- Department of Mathematics, The College of Saint Rose, Albany, NY 12203, USA
| | - Mark J. Embrechts
- Department of Industrial and Systems Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Uwe Kruger
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
11
|
Satoh AO, Fujioka Y, Kashiwagi S, Yoshida A, Fujioka M, Sasajima H, Nanbo A, Amano M, Ohba Y. Interaction between PI3K and the VDAC2 channel tethers Ras-PI3K-positive endosomes to mitochondria and promotes endosome maturation. Cell Rep 2023; 42:112229. [PMID: 36906852 DOI: 10.1016/j.celrep.2023.112229] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/01/2023] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
Intracellular organelles of mammalian cells communicate with one another during various cellular processes. The functions and molecular mechanisms of such interorganelle association remain largely unclear, however. We here identify voltage-dependent anion channel 2 (VDAC2), a mitochondrial outer membrane protein, as a binding partner of phosphoinositide 3-kinase (PI3K), a regulator of clathrin-independent endocytosis downstream of the small GTPase Ras. VDAC2 tethers endosomes positive for the Ras-PI3K complex to mitochondria in response to cell stimulation with epidermal growth factor and promotes clathrin-independent endocytosis, as well as endosome maturation at membrane association sites. With an optogenetics system to induce mitochondrion-endosome association, we find that, in addition to its structural role in such association, VDAC2 is functionally implicated in the promotion of endosome maturation. The mitochondrion-endosome association thus plays a role in the regulation of clathrin-independent endocytosis and endosome maturation.
Collapse
Affiliation(s)
- Aya O Satoh
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Kita-ku, Sapporo 060-8638, Japan
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan; Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-0812, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Kita-ku, Sapporo 060-8638, Japan
| | - Sayaka Kashiwagi
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan; Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-0812, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Kita-ku, Sapporo 060-8638, Japan
| | - Aiko Yoshida
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan; Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-0812, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Kita-ku, Sapporo 060-8638, Japan
| | - Mari Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Kita-ku, Sapporo 060-8638, Japan
| | - Hitoshi Sasajima
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Asuka Nanbo
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Maho Amano
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Kita-ku, Sapporo 060-8638, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan; Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-0812, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Kita-ku, Sapporo 060-8638, Japan.
| |
Collapse
|
12
|
Wu X, Zhu H, Xu Y, Kong B, Tan Q. Chronic wounds: pathological characteristics and their stem cell-based therapies. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
13
|
Progress in mesenchymal stem cell mitochondria transfer for the repair of tissue injury and treatment of disease. Biomed Pharmacother 2022; 153:113482. [DOI: 10.1016/j.biopha.2022.113482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/19/2022] Open
|
14
|
Gayan S, Joshi G, Dey T. Biomarkers of mitochondrial origin: a futuristic cancer diagnostic. Integr Biol (Camb) 2022; 14:77-88. [PMID: 35780307 DOI: 10.1093/intbio/zyac008] [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: 12/20/2021] [Revised: 05/17/2022] [Accepted: 05/27/2022] [Indexed: 11/12/2022]
Abstract
Cancer is a highly fatal disease without effective early-stage diagnosis and proper treatment. Along with the oncoproteins and oncometabolites, several organelles from cancerous cells are also emerging as potential biomarkers. Mitochondria isolated from cancer cells are one such biomarker candidates. Cancerous mitochondria exhibit different profiles compared with normal ones in morphology, genomic, transcriptomic, proteomic and metabolic landscape. Here, the possibilities of exploring such characteristics as potential biomarkers through single-cell omics and Artificial Intelligence (AI) are discussed. Furthermore, the prospects of exploiting the biomarker-based diagnosis and its futuristic utilization through circulatory tumor cell technology are analyzed. A successful alliance of circulatory tumor cell isolation protocols and a single-cell omics platform can emerge as a next-generation diagnosis and personalized treatment procedure.
Collapse
Affiliation(s)
- Sukanya Gayan
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Gargee Joshi
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Tuli Dey
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
15
|
Kalyanaraman B. Exploiting the tumor immune microenvironment and immunometabolism using mitochondria-targeted drugs: Challenges and opportunities in racial disparity and cancer outcome research. FASEB J 2022; 36:e22226. [PMID: 35233843 PMCID: PMC9242412 DOI: 10.1096/fj.202101862r] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 12/16/2022]
Abstract
Black and Hispanic cancer patients have a higher incidence of cancer mortality. Many factors (e.g., socioeconomic differences, insufficient access to healthcare) contribute to racial disparity. Emerging research implicates biological disparity in cancer outcomes. Studies show distinct differences in the tumor immune microenvironment (TIME) in Black cancer patients. Studies also have linked altered mitochondrial metabolism to changes in immune cell activation in TIME. Recent publications revealed a novel immunomodulatory role for triphenylphosphonium-based mitochondrial-targeted drugs (MTDs). These are synthetically modified, naturally occurring molecules (e.g., honokiol, magnolol, metformin) or FDA-approved small molecule drugs (e.g., atovaquone, hydroxyurea). Modifications involve conjugating the parent molecule via an alkyl linker chain to a triphenylphosphonium moiety. These modified molecules (e.g., Mito-honokiol, Mito-magnolol, Mito-metformin, Mito-atovaquone, Mito-hydroxyurea) accumulate in tumor cell mitochondria more effectively than in normal cells and inhibit mitochondrial respiration, induce reactive oxygen species, activate AMPK and redox transcription factors, and inhibit cancer cell proliferation. Besides these intrinsic effects of MTDs in redox signaling and proliferation in tumors, MTDs induced extrinsic effects in the TIME of mouse xenografts. MTD treatment inhibited tumor-suppressive immune cells, myeloid-derived suppressor cells, and regulatory T cells, and activated T cells and antitumor immune effects. One key biological disparity in Black cancer patients was related to altered mitochondrial oxidative metabolism; MTDs targeting vulnerabilities in tumor cells and the TIME may help us understand this biological disparity. Clinical trials should include an appropriate number of Black and Hispanic cancer patients and should validate the intratumoral, antihypoxic effects of MTDs with imaging.
Collapse
Affiliation(s)
- Balaraman Kalyanaraman
- Department of BiophysicsMedical College of WisconsinMilwaukeeWisconsinUSA
- Center for Disease Prevention ResearchMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
16
|
Mapuskar KA, Steinbach EJ, Zaher A, Riley DP, Beardsley RA, Keene JL, Holmlund JT, Anderson CM, Zepeda-Orozco D, Buatti JM, Spitz DR, Allen BG. Mitochondrial Superoxide Dismutase in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2021; 10:antiox10091329. [PMID: 34572961 PMCID: PMC8469643 DOI: 10.3390/antiox10091329] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is a chemotherapy agent commonly used to treat a wide variety of cancers. Despite the potential for both severe acute and chronic side effects, it remains a preferred therapeutic option for many malignancies due to its potent anti-tumor activity. Common cisplatin-associated side-effects include acute kidney injury (AKI) and chronic kidney disease (CKD). These renal injuries may cause delays and potentially cessation of cisplatin therapy and have long-term effects on renal function reserve. Thus, developing mechanism-based interventional strategies that minimize cisplatin-associated kidney injury without reducing efficacy would be of great benefit. In addition to its action of cross-linking DNA, cisplatin has been shown to affect mitochondrial metabolism, resulting in mitochondrially derived reactive oxygen species (ROS). Increased ROS formation in renal proximal convoluted tubule cells is associated with cisplatin-induced AKI and CKD. We review the mechanisms by which cisplatin may induce AKI and CKD and discuss the potential of mitochondrial superoxide dismutase mimetics to prevent platinum-associated nephrotoxicity.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Emily J. Steinbach
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Amira Zaher
- Biomedical Science Program, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA;
| | - Dennis P. Riley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Robert A. Beardsley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jeffery L. Keene
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jon T. Holmlund
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Carryn M. Anderson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Diana Zepeda-Orozco
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Nephrology, Department of Pediatrics, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - John M. Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
- Correspondence: ; Tel.: +1-319-335-8019; Fax: +1-319-335-8039
| |
Collapse
|
17
|
Machine learning-based classification of mitochondrial morphology in primary neurons and brain. Sci Rep 2021; 11:5133. [PMID: 33664336 PMCID: PMC7933342 DOI: 10.1038/s41598-021-84528-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/17/2021] [Indexed: 01/31/2023] Open
Abstract
The mitochondrial network continually undergoes events of fission and fusion. Under physiologic conditions, the network is in equilibrium and is characterized by the presence of both elongated and punctate mitochondria. However, this balanced, homeostatic mitochondrial profile can change morphologic distribution in response to various stressors. Therefore, it is imperative to develop a method that robustly measures mitochondrial morphology with high accuracy. Here, we developed a semi-automated image analysis pipeline for the quantitation of mitochondrial morphology for both in vitro and in vivo applications. The image analysis pipeline was generated and validated utilizing images of primary cortical neurons from transgenic mice, allowing genetic ablation of key components of mitochondrial dynamics. This analysis pipeline was further extended to evaluate mitochondrial morphology in vivo through immunolabeling of brain sections as well as serial block-face scanning electron microscopy. These data demonstrate a highly specific and sensitive method that accurately classifies distinct physiological and pathological mitochondrial morphologies. Furthermore, this workflow employs the use of readily available, free open-source software designed for high throughput image processing, segmentation, and analysis that is customizable to various biological models.
Collapse
|
18
|
Kesisova IA, Robinson BP, Spiliotis ET. A septin GTPase scaffold of dynein-dynactin motors triggers retrograde lysosome transport. J Cell Biol 2021; 220:211663. [PMID: 33416861 PMCID: PMC7802366 DOI: 10.1083/jcb.202005219] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/22/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
The metabolic and signaling functions of lysosomes depend on their intracellular positioning and trafficking, but the underlying mechanisms are little understood. Here, we have discovered a novel septin GTPase-based mechanism for retrograde lysosome transport. We found that septin 9 (SEPT9) associates with lysosomes, promoting the perinuclear localization of lysosomes in a Rab7-independent manner. SEPT9 targeting to mitochondria and peroxisomes is sufficient to recruit dynein and cause perinuclear clustering. We show that SEPT9 interacts with both dynein and dynactin through its GTPase domain and N-terminal extension, respectively. Strikingly, SEPT9 associates preferentially with the dynein intermediate chain (DIC) in its GDP-bound state, which favors dimerization and assembly into septin multimers. In response to oxidative cell stress induced by arsenite, SEPT9 localization to lysosomes is enhanced, promoting the perinuclear clustering of lysosomes. We posit that septins function as GDP-activated scaffolds for the cooperative assembly of dynein-dynactin, providing an alternative mechanism of retrograde lysosome transport at steady state and during cellular adaptation to stress.
Collapse
|
19
|
Mito Hacker: a set of tools to enable high-throughput analysis of mitochondrial network morphology. Sci Rep 2020; 10:18941. [PMID: 33144635 PMCID: PMC7642274 DOI: 10.1038/s41598-020-75899-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are highly dynamic organelles that can exhibit a wide range of morphologies. Mitochondrial morphology can differ significantly across cell types, reflecting different physiological needs, but can also change rapidly in response to stress or the activation of signaling pathways. Understanding both the cause and consequences of these morphological changes is critical to fully understanding how mitochondrial function contributes to both normal and pathological physiology. However, while robust and quantitative analysis of mitochondrial morphology has become increasingly accessible, there is a need for new tools to generate and analyze large data sets of mitochondrial images in high throughput. The generation of such datasets is critical to fully benefit from rapidly evolving methods in data science, such as neural networks, that have shown tremendous value in extracting novel biological insights and generating new hypotheses. Here we describe a set of three computational tools, Cell Catcher, Mito Catcher and MiA, that we have developed to extract extensive mitochondrial network data on a single-cell level from multi-cell fluorescence images. Cell Catcher automatically separates and isolates individual cells from multi-cell images; Mito Catcher uses the statistical distribution of pixel intensities across the mitochondrial network to detect and remove background noise from the cell and segment the mitochondrial network; MiA uses the binarized mitochondrial network to perform more than 100 mitochondria-level and cell-level morphometric measurements. To validate the utility of this set of tools, we generated a database of morphological features for 630 individual cells that encode 0, 1 or 2 alleles of the mitochondrial fission GTPase Drp1 and demonstrate that these mitochondrial data could be used to predict Drp1 genotype with 87% accuracy. Together, this suite of tools enables the high-throughput and automated collection of detailed and quantitative mitochondrial structural information at a single-cell level. Furthermore, the data generated with these tools, when combined with advanced data science approaches, can be used to generate novel biological insights.
Collapse
|
20
|
Caveolin-1-mediated sphingolipid oncometabolism underlies a metabolic vulnerability of prostate cancer. Nat Commun 2020; 11:4279. [PMID: 32855410 PMCID: PMC7453025 DOI: 10.1038/s41467-020-17645-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
Plasma and tumor caveolin-1 (Cav-1) are linked with disease progression in prostate cancer. Here we report that metabolomic profiling of longitudinal plasmas from a prospective cohort of 491 active surveillance (AS) participants indicates prominent elevations in plasma sphingolipids in AS progressors that, together with plasma Cav-1, yield a prognostic signature for disease progression. Mechanistic studies of the underlying tumor supportive onco-metabolism reveal coordinated activities through which Cav-1 enables rewiring of cancer cell lipid metabolism towards a program of 1) exogenous sphingolipid scavenging independent of cholesterol, 2) increased cancer cell catabolism of sphingomyelins to ceramide derivatives and 3) altered ceramide metabolism that results in increased glycosphingolipid synthesis and efflux of Cav-1-sphingolipid particles containing mitochondrial proteins and lipids. We also demonstrate, using a prostate cancer syngeneic RM-9 mouse model and established cell lines, that this Cav-1-sphingolipid program evidences a metabolic vulnerability that is targetable to induce lethal mitophagy as an anti-tumor therapy.
Collapse
|
21
|
Cadmium elicits alterations in mitochondrial morphology and functionality in C3H10T1/2Cl8 mouse embryonic fibroblasts. Biochim Biophys Acta Gen Subj 2020; 1864:129568. [DOI: 10.1016/j.bbagen.2020.129568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/28/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022]
|
22
|
Ahmed MU, Velkov T, Zhou QT, Fulcher AJ, Callaghan J, Zhou F, Chan K, Azad MAK, Li J. Intracellular localization of polymyxins in human alveolar epithelial cells. J Antimicrob Chemother 2020; 74:48-57. [PMID: 30357331 DOI: 10.1093/jac/dky409] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/06/2018] [Indexed: 01/05/2023] Open
Abstract
Background Current inhaled polymyxin therapy is empirical and often large doses are administered, which can lead to pulmonary adverse effects. There is a dearth of information on the mechanisms of polymyxin-induced lung toxicity and their intracellular localization in lung epithelial cells. Objectives To investigate the intracellular localization of polymyxins in human lung epithelial A549 cells. Methods A549 cells were treated with polymyxin B and intracellular organelles (early and late endosomes, endoplasmic reticulum, mitochondria, lysosomes and autophagosomes), ubiquitin protein and polymyxin B were visualized using immunostaining and confocal microscopy. Fluorescence intensities of the organelles and polymyxin B were quantified and correlated for co-localization using ImageJ and Imaris platforms. Results Polymyxin B co-localized with early endosomes, lysosomes and ubiquitin at 24 h. Significantly increased lysosomal activity and the autophagic protein LC3A were observed after 0.5 and 1.0 mM polymyxin B treatment at 24 h. Polymyxin B also significantly co-localized with mitochondria (Pearson's R = 0.45) and led to the alteration of mitochondrial morphology from filamentous to fragmented form (n = 3, P < 0.001). These results are in line with the polymyxin-induced activation of the mitochondrial apoptotic pathway observed in A549 cells. Conclusions Accumulation of polymyxins on mitochondria probably caused mitochondrial toxicity, resulting in increased oxidative stress and cell death. The formation of autophagosomes and lysosomes was likely a cellular response to the polymyxin-induced stress and played a defensive role by disassembling dysfunctional organelles and proteins. Our study provides new mechanistic information on polymyxin-induced lung toxicity, which is vital for optimizing inhaled polymyxins in the clinic.
Collapse
Affiliation(s)
- Maizbha U Ahmed
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.,Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Victoria, Australia
| | - Tony Velkov
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Victoria, Australia
| | - Judy Callaghan
- Monash Micro Imaging, Monash University, Victoria, Australia
| | - Fanfan Zhou
- School of Pharmacy, The University of Sydney, Camperdown, NSW, Australia
| | - Kim Chan
- School of Pharmacy, The University of Sydney, Camperdown, NSW, Australia
| | - Mohammad A K Azad
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Victoria, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Victoria, Australia
| |
Collapse
|
23
|
Mirzapoiazova T, Li H, Nathan A, Srivstava S, Nasser MW, Lennon F, Armstrong B, Mambetsariev I, Chu PG, Achuthan S, Batra SK, Kulkarni P, Salgia R. Monitoring and Determining Mitochondrial Network Parameters in Live Lung Cancer Cells. J Clin Med 2019; 8:jcm8101723. [PMID: 31635288 PMCID: PMC6832496 DOI: 10.3390/jcm8101723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are dynamic organelles that constantly fuse and divide, forming dynamic tubular networks. Abnormalities in mitochondrial dynamics and morphology are linked to diverse pathological states, including cancer. Thus, alterations in mitochondrial parameters could indicate early events of disease manifestation or progression. However, finding reliable and quantitative tools for monitoring mitochondria and determining the network parameters, particularly in live cells, has proven challenging. Here, we present a 2D confocal imaging-based approach that combines automatic mitochondrial morphology and dynamics analysis with fractal analysis in live small cell lung cancer (SCLC) cells. We chose SCLC cells as a test case since they typically have very little cytoplasm, but an abundance of smaller mitochondria compared to many of the commonly used cell types. The 2D confocal images provide a robust approach to quantitatively measure mitochondrial dynamics and morphology in live cells. Furthermore, we performed 3D reconstruction of electron microscopic images and show that the 3D reconstruction of the electron microscopic images complements this approach to yield better resolution. The data also suggest that the parameters of mitochondrial dynamics and fractal dimensions are sensitive indicators of cellular response to subtle perturbations, and hence, may serve as potential markers of drug response in lung cancer.
Collapse
Affiliation(s)
- Tamara Mirzapoiazova
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Haiqing Li
- Center for Informatics, City of Hope National Medical Center, Duarte, CA 91010, USA.
- Department of Computational & Quantitative Medicine, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010, USA.
| | - Anusha Nathan
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Saumya Srivstava
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Mohd W Nasser
- University of Nebraska, Medical Center, Nebraska, NE 68198, USA.
| | | | - Brian Armstrong
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Peiguo G Chu
- Department of Anatomic Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Srisairam Achuthan
- Center for Informatics, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Surinder K Batra
- University of Nebraska, Medical Center, Nebraska, NE 68198, USA.
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
24
|
Matthias J, Kanagasundaram T, Kopka K, Kramer CS. Synthesis of a dihalogenated pyridinyl silicon rhodamine for mitochondrial imaging by a halogen dance rearrangement. Beilstein J Org Chem 2019; 15:2333-2343. [PMID: 31666868 PMCID: PMC6808212 DOI: 10.3762/bjoc.15.226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/05/2019] [Indexed: 12/30/2022] Open
Abstract
Background: Since their first synthesis, silicon xanthenes and the subsequently developed silicon rhodamines (SiR) gained a lot of attention as attractive fluorescence dyes offering a broad field of application. We aimed for the synthesis of a fluorinable pyridinyl silicon rhodamine for the use in multimodal (PET/OI) medical imaging of mitochondria in cancerous cells. Results: A dihalogenated fluorinatable pyridinyl rhodamine could be successfully synthesized with the high yield of 85% by application of a halogen dance (HD) rearrangement. The near-infrared dye shows a quantum yield of 0.34, comparable to other organelle targeting SiR derivatives and absorbs at 665 nm (εmax = 34 000 M−1cm−1) and emits at 681 nm (τ = 1.9 ns). Using colocalization experiments with MitoTracker® Green FM, we could prove the intrinsic targeting ability to mitochondria in two human cell lines (Pearson coefficient >0.8). The dye is suitable for live cell STED nanoscopy imaging and shows a nontoxic profile which makes it an appropriate candidate for medical imaging. Conclusions: We present a biocompatible, nontoxic, small molecule near-infrared dye with the option of subsequent radiolabelling and excellent optical properties for medical and bioimaging. As a compound with intrinsic mitochondria targeting ability, the radiolabelled analogue can be applied in multimodal (PET/OI) imaging of mitochondria for diagnostic and therapeutic use in, e.g., cancer patients.
Collapse
Affiliation(s)
- Jessica Matthias
- Max Planck Institute for Medical Research, Department of Optical Nanoscopy, Jahnstraße 29, 69120 Heidelberg, Germany.,Helmholtz International Graduate School, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 223, 69120 Heidelberg, Germany
| | - Thines Kanagasundaram
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 223, 69120 Heidelberg, Germany.,Institute of Inorganic Chemistry, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 223, 69120 Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Carsten S Kramer
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 223, 69120 Heidelberg, Germany
| |
Collapse
|
25
|
Force Spectrum Microscopy Using Mitochondrial Fluctuations of Control and ATP-Depleted Cells. Biophys J 2019; 114:2933-2944. [PMID: 29925029 DOI: 10.1016/j.bpj.2018.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/15/2018] [Accepted: 05/01/2018] [Indexed: 11/24/2022] Open
Abstract
A single-cell assay of active and passive intracellular mechanical properties of mammalian cells could give significant insight into cellular processes. Force spectrum microscopy (FSM) is one such technique, which combines the spontaneous motion of probe particles and the mechanical properties of the cytoskeleton measured by active microrheology using optical tweezers to determine the force spectrum of the cytoskeleton. A simpler and noninvasive method to perform FSM would be very useful, enabling its widespread adoption. Here, we develop an alternative method of FSM using measurement of the fluctuating motion of mitochondria. Mitochondria of the C3H-10T1/2 cell line were labeled and tracked using confocal microscopy. Mitochondrial probes were selected based on morphological characteristics, and their mean-square displacement, creep compliance, and distributions of directional change were measured. We found that the creep compliance of mitochondria resembles that of particles in viscoelastic media. However, comparisons of creep compliance between controls and cells treated with pharmacological agents showed that perturbations to the actomysoin network had surprisingly small effects on mitochondrial fluctuations, whereas microtubule disruption and ATP depletion led to a significantly decreased creep compliance. We used properties of the distribution of directional change to identify a regime of thermally dominated fluctuations in ATP-depleted cells, allowing us to estimate the viscoelastic parameters for a range of timescales. We then determined the force spectrum by combining these viscoelastic properties with measurements of spontaneous fluctuations tracked in control cells. Comparisons with previous measurements made using FSM revealed an excellent match.
Collapse
|
26
|
Mitochondria in the signaling pathways that control longevity and health span. Ageing Res Rev 2019; 54:100940. [PMID: 31415807 PMCID: PMC7479635 DOI: 10.1016/j.arr.2019.100940] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/09/2019] [Accepted: 08/06/2019] [Indexed: 12/26/2022]
Abstract
Genetic and pharmacological intervention studies have identified evolutionarily conserved and functionally interconnected networks of cellular energy homeostasis, nutrient-sensing, and genome damage response signaling pathways, as prominent regulators of longevity and health span in various species. Mitochondria are the primary sites of ATP production and are key players in several other important cellular processes. Mitochondrial dysfunction diminishes tissue and organ functional performance and is a commonly considered feature of the aging process. Here we review the evidence that through reciprocal and multilevel functional interactions, mitochondria are implicated in the lifespan modulation function of these pathways, which altogether constitute a highly dynamic and complex system that controls the aging process. An important characteristic of these pathways is their extensive crosstalk and apparent malleability to modification by non-invasive pharmacological, dietary, and lifestyle interventions, with promising effects on lifespan and health span in animal models and potentially also in humans.
Collapse
|
27
|
Zhang Z, Chen L, Humphries B, Brien R, Wicha MS, Luker KE, Luker GD, Chen YC, Yoon E. Morphology-based prediction of cancer cell migration using an artificial neural network and a random decision forest. Integr Biol (Camb) 2019; 10:758-767. [PMID: 30420987 DOI: 10.1039/c8ib00106e] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metastasis is the cause of death in most patients of breast cancer and other solid malignancies. Identification of cancer cells with highly migratory capability to metastasize relies on markers for epithelial-to-mesenchymal transition (EMT), a process increasing cell migration and metastasis. Marker-based approaches are limited by inconsistences among patients, types of cancer, and partial EMT states. Alternatively, we analyzed cancer cell migration behavior using computer vision. Using a microfluidic single-cell migration chip and high-content imaging, we extracted morphological features and recorded migratory direction and speed of breast cancer cells. By applying a Random Decision Forest (RDF) and an Artificial Neural Network (ANN), we achieved over 99% accuracy for cell movement direction prediction and 91% for speed prediction. Unprecedentedly, we identified highly motile cells and non-motile cells based on microscope images and a machine learning model, and pinpointed and validated morphological features determining cell migration, including not only known features related to cell polarization but also novel ones that can drive future mechanistic studies. Predicting cell movement by computer vision and machine learning establishes a ground-breaking approach to analyze cell migration and metastasis.
Collapse
Affiliation(s)
- Zhixiong Zhang
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
The Mitochondrion as an Emerging Therapeutic Target in Cancer. Trends Mol Med 2019; 26:119-134. [PMID: 31327706 DOI: 10.1016/j.molmed.2019.06.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022]
Abstract
Mitochondria have emerged as important pharmacological targets because of their key role in cellular proliferation and death. In tumor tissues, mitochondria can switch metabolic phenotypes to meet the challenges of high energy demand and macromolecular synthesis. Furthermore, mitochondria can engage in crosstalk with the tumor microenvironment, and signals from cancer-associated fibroblasts can impinge on mitochondria. Cancer cells can also acquire a hybrid phenotype in which both glycolysis and oxidative phosphorylation (OXPHOS) can be utilized. This hybrid phenotype can facilitate metabolic plasticity of cancer cells more specifically in metastasis and therapy-resistance. In light of the metabolic heterogeneity and plasticity of cancer cells that had until recently remained unappreciated, strategies targeting cancer metabolic dependency appear to be promising in the development of novel and effective cancer therapeutics.
Collapse
|
29
|
Su X, Yuan T, Wang Z, Song K, Li R, Yuan C, Kong B. Two-Dimensional Light Scattering Anisotropy Cytometry for Label-Free Classification of Ovarian Cancer Cells via Machine Learning. Cytometry A 2019; 97:24-30. [PMID: 31313517 DOI: 10.1002/cyto.a.23865] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 06/14/2019] [Accepted: 07/01/2019] [Indexed: 12/24/2022]
Abstract
We develop a single-mode fiber-based cytometer for the obtaining of two-dimensional (2D) light scattering patterns from static single cells. Anisotropy of the 2D light scattering patterns of single cells from ovarian cancer and normal cell lines is investigated by histograms of oriented gradients (HOG) method. By analyzing the HOG descriptors with support vector machine, an accuracy rate of 92.84% is achieved for the automatic classification of these two kinds of label-free cells. The 2D light scattering anisotropy cytometry combined with machine learning may provide a label-free, automatic method for screening of ovarian cancer cells, and other types of cells. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Xuantao Su
- Institute of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Tao Yuan
- Institute of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Zhiwen Wang
- Institute of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Kun Song
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Rongrong Li
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Cunzhong Yuan
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, China
| |
Collapse
|
30
|
Sarmiento-Salinas FL, Delgado-Magallón A, Montes-Alvarado JB, Ramírez-Ramírez D, Flores-Alonso JC, Cortés-Hernández P, Reyes-Leyva J, Herrera-Camacho I, Anaya-Ruiz M, Pelayo R, Millán-Pérez-Peña L, Maycotte P. Breast Cancer Subtypes Present a Differential Production of Reactive Oxygen Species (ROS) and Susceptibility to Antioxidant Treatment. Front Oncol 2019; 9:480. [PMID: 31231612 PMCID: PMC6568240 DOI: 10.3389/fonc.2019.00480] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022] Open
Abstract
Due to their crucial role in cell metabolism and homeostasis, alterations in mitochondrial biology and function have been related to the progression of diverse diseases including cancer. One of the consequences associated to mitochondrial dysfunction is the production of reactive oxygen species (ROS). ROS are known to have a controversial role during cancer initiation and progression and although several studies have tried to manipulate intracellular ROS levels using antioxidants or pro-oxidation conditions, it is not yet clear how to target oxidation for cancer therapy. In this study, we found differences in mitochondrial morphology in breast cancer cells when compared to a non-tumorigenic cell line and differences in mitochondrial function among breast cancer subtypes when exploring gene-expression data from the TCGA tumor dataset. Interestingly, we found increased ROS levels in triple negative breast cancer (TNBC) cell lines and a dependency on ROS for survival since antioxidant treatment induced cell death in TNBC cells but not in an estrogen receptor positive (ER+) cell line. Moreover, we identified the mitochondria as the main source of ROS in TNBC cell lines. Our results indicate a potential use for ROS as a target for therapy in the TNBC subtype which currently has the worst prognosis among all breast cancers and remains as the only breast cancer subtype which lacks a targeted therapy.
Collapse
Affiliation(s)
- Fabiola Lilí Sarmiento-Salinas
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico.,Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alam Delgado-Magallón
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico.,Departamento de Bioquímica, Instituto Tecnológico de Acapulco, Acapulco de Juárez, Mexico
| | | | - Dalia Ramírez-Ramírez
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | | | - Paulina Cortés-Hernández
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Julio Reyes-Leyva
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Irma Herrera-Camacho
- Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Maricruz Anaya-Ruiz
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Rosana Pelayo
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | | | - Paola Maycotte
- CONACYT-Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Puebla, Mexico
| |
Collapse
|
31
|
Naviaux RK. Incomplete Healing as a Cause of Aging: The Role of Mitochondria and the Cell Danger Response. BIOLOGY 2019; 8:biology8020027. [PMID: 31083530 PMCID: PMC6627909 DOI: 10.3390/biology8020027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
Abstract
The rate of biological aging varies cyclically and episodically in response to changing environmental conditions and the developmentally-controlled biological systems that sense and respond to those changes. Mitochondria and metabolism are fundamental regulators, and the cell is the fundamental unit of aging. However, aging occurs at all anatomical levels. At levels above the cell, aging in different tissues is qualitatively, quantitatively, and chronologically distinct. For example, the heart can age faster and differently than the kidney and vice versa. Two multicellular features of aging that are universal are: (1) a decrease in physiologic reserve capacity, and (2) a decline in the functional communication between cells and organ systems, leading to death. Decreases in reserve capacity and communication impose kinetic limits on the rate of healing after new injuries, resulting in dyssynchronous and incomplete healing. Exercise mitigates against these losses, but recovery times continue to increase with age. Reinjury before complete healing results in the stacking of incomplete cycles of healing. Developmentally delayed and arrested cells accumulate in the three stages of the cell danger response (CDR1, 2, and 3) that make up the healing cycle. Cells stuck in the CDR create physical and metabolic separation—buffer zones of reduced communication—between previously adjoining, synergistic, and metabolically interdependent cells. Mis-repairs and senescent cells accumulate, and repeated iterations of incomplete cycles of healing lead to progressively dysfunctional cellular mosaics in aging tissues. Metabolic cross-talk between mitochondria and the nucleus, and between neighboring and distant cells via signaling molecules called metabokines regulates the completeness of healing. Purinergic signaling and sphingolipids play key roles in this process. When viewed against the backdrop of the molecular features of the healing cycle, the incomplete healing model provides a new framework for understanding the hallmarks of aging and generates a number of testable hypotheses for new treatments.
Collapse
Affiliation(s)
- Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, Departments of Medicine, Pediatrics, Pathology, University of California, San Diego School of Medicine, San Diego, CA 92103, USA.
| |
Collapse
|
32
|
Venkatachalapathy M, Belapurkar V, Jose M, Gautier A, Nair D. Live cell super resolution imaging by radial fluctuations using fluorogen binding tags. NANOSCALE 2019; 11:3626-3632. [PMID: 30734810 DOI: 10.1039/c8nr07809b] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Fluorescence-Activating and absorption-Shifting Tag (FAST) is a novel genetically encoded optical highlighter probe. Since the fluorescence of FAST originates from the stochastic and reversible diffusive association of a fluorogenic ligand, we investigate the application of FAST using Super-Resolution Radial Fluctuations (SRRF) to achieve routine imaging below the diffraction limit in a widefield epifluorescence microscope. We show that intensity fluctuation analysis like SRRF allows the imaging of FAST-tagged proteins with sub - 100 nm resolution in live cells. FAST co-labeled with conventional fluorophores enables real time multicolour 2D and 3D super-resolution imaging, indicating that FAST can be used for the observation of sub-diffraction limited structures in both living and fixed samples.
Collapse
|
33
|
Sung J, Rho JG, Jeon GG, Chu Y, Min JS, Lee S, Kim JH, Kim W, Kim E. A New Infrared Probe Targeting Mitochondria via Regulation of Molecular Hydrophobicity. Bioconjug Chem 2019; 30:210-217. [PMID: 30562008 DOI: 10.1021/acs.bioconjchem.8b00845] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Herein, we developed a near-infrared (NIR) fluorescent probe for mitochondrial staining based on the NIR fluorochrome, silicon-rhodamine. The hydrophobicity of the fluorescent core was systematically modified by conjugation with 10 different commercial amines. The resulting fluorescent compounds exhibited similar photophysical properties but diverse hydrophobicity. We identified the optimal level of hydrophobicity associated with high mitochondrial targeting efficiency. In particular, the SiR-Mito 8 probe provided excellent mitochondrial staining and successfully differentiated the live Hep3B cancer cells from normal L02 cells in vitro.
Collapse
Affiliation(s)
| | | | | | - Yeonjeong Chu
- Center for Neuro-Medicine, Brain Science Institute , Korea Institute of Science and Technology , Seoul 02792 , Korea
| | | | - Sanghee Lee
- Center for Neuro-Medicine, Brain Science Institute , Korea Institute of Science and Technology , Seoul 02792 , Korea
| | | | | | | |
Collapse
|
34
|
Abstract
Mitochondria are dynamic organelles that in most cells behave as a dynamic network and can change their biogenesis and structure depending on the cell needs or as a response to different conditions. Analyzing the architecture of mitochondria is determinant to describe their state and function. In this chapter, image processing techniques are applied in a workflow manner to segment the mitochondrial network and extract the most relevant parameters that enable an accurate morphology analysis. This workflow is programmed with ImageJ macro language and can be applied to automatically analyze multiple cells from multiple images or tiles. When combined with multiwell plates and automated microscopy, this method may allow to perform high content image analysis of hundreds of cells under different conditions.
Collapse
Affiliation(s)
- Anna Bosch
- Scientific and Technological Centers (CCiTUB), Universitat de Barcelona, Barcelona, Spain
| | - Maria Calvo
- Advanced Optical Microscopy Facility, Scientific and Technological Centers of University of Barcelona, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
35
|
Newell C, Sabouny R, Hittel DS, Shutt TE, Khan A, Klein MS, Shearer J. Mesenchymal Stem Cells Shift Mitochondrial Dynamics and Enhance Oxidative Phosphorylation in Recipient Cells. Front Physiol 2018; 9:1572. [PMID: 30555336 PMCID: PMC6282049 DOI: 10.3389/fphys.2018.01572] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are the most commonly used cells in tissue engineering and regenerative medicine. MSCs can promote host tissue repair through several different mechanisms including donor cell engraftment, release of cell signaling factors, and the transfer of healthy organelles to the host. In the present study, we examine the specific impacts of MSCs on mitochondrial morphology and function in host tissues. Employing in vitro cell culture of inherited mitochondrial disease and an in vivo animal experimental model of low-grade inflammation (high fat feeding), we show human-derived MSCs to alter mitochondrial function. MSC co-culture with skin fibroblasts from mitochondrial disease patients rescued aberrant mitochondrial morphology from a fission state to a more fused appearance indicating an effect of MSC co-culture on host cell mitochondrial network formation. In vivo experiments confirmed mitochondrial abundance and mitochondrial oxygen consumption rates were elevated in host tissues following MSC treatment. Furthermore, microarray profiling identified 226 genes with differential expression in the liver of animals treated with MSC, with cellular signaling, and actin cytoskeleton regulation as key upregulated processes. Collectively, our data indicate that MSC therapy rescues impaired mitochondrial morphology, enhances host metabolic capacity, and induces widespread host gene shifting. These results highlight the potential of MSCs to modulate mitochondria in both inherited and pathological disease states.
Collapse
Affiliation(s)
- Christopher Newell
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Rasha Sabouny
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Dustin S Hittel
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Timothy E Shutt
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aneal Khan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Departments of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Matthias S Klein
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Jane Shearer
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
36
|
Zahedi A, On V, Phandthong R, Chaili A, Remark G, Bhanu B, Talbot P. Deep Analysis of Mitochondria and Cell Health Using Machine Learning. Sci Rep 2018; 8:16354. [PMID: 30397207 PMCID: PMC6218515 DOI: 10.1038/s41598-018-34455-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 10/16/2018] [Indexed: 12/22/2022] Open
Abstract
There is a critical need for better analytical methods to study mitochondria in normal and diseased states. Mitochondrial image analysis is typically done on still images using slow manual methods or automated methods of limited types of features. MitoMo integrated software overcomes these bottlenecks by automating rapid unbiased quantitative analysis of mitochondrial morphology, texture, motion, and morphogenesis and advances machine-learning classification to predict cell health by combining features. Our pixel-based approach for motion analysis evaluates the magnitude and direction of motion of: (1) molecules within mitochondria, (2) individual mitochondria, and (3) distinct morphological classes of mitochondria. MitoMo allows analysis of mitochondrial morphogenesis in time-lapse videos to study early progression of cellular stress. Biological applications are presented including: (1) establishing normal phenotypes of mitochondria in different cell types; (2) quantifying stress-induced mitochondrial hyperfusion in cells treated with an environmental toxicant, (3) tracking morphogenesis in mitochondria undergoing swelling, and (4) evaluating early changes in cell health when morphological abnormalities are not apparent. MitoMo unlocks new information on mitochondrial phenotypes and dynamics by enabling deep analysis of mitochondrial features in any cell type and can be applied to a broad spectrum of research problems in cell biology, drug testing, toxicology, and medicine.
Collapse
Affiliation(s)
- Atena Zahedi
- Graduate Program in Bioengineering, University of California, Riverside, CA., USA
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA., USA
| | - Vincent On
- Department of Electrical & Computer Engineering, University of California, Riverside, CA., USA
| | - Rattapol Phandthong
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA., USA
| | - Angela Chaili
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA., USA
| | - Guadalupe Remark
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA., USA
| | - Bir Bhanu
- Graduate Program in Bioengineering, University of California, Riverside, CA., USA
- Department of Electrical & Computer Engineering, University of California, Riverside, CA., USA
- Department of Computer Science, University of California, Riverside, CA., USA
| | - Prue Talbot
- Graduate Program in Bioengineering, University of California, Riverside, CA., USA.
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA., USA.
| |
Collapse
|
37
|
Kalainayakan SP, FitzGerald KE, Konduri PC, Vidal C, Zhang L. Essential roles of mitochondrial and heme function in lung cancer bioenergetics and tumorigenesis. Cell Biosci 2018; 8:56. [PMID: 30410721 PMCID: PMC6215344 DOI: 10.1186/s13578-018-0257-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 10/26/2018] [Indexed: 01/12/2023] Open
Abstract
Contrary to Warburg’s hypothesis, mitochondrial oxidative phosphorylation (OXPHOS) contributes significantly to fueling cancer cells. Several recent studies have demonstrated that radiotherapy-resistant and chemotherapy-resistant cancer cells depend on OXPHOS for survival and progression. Several cancers exhibit an increased risk in association with heme intake. Mitochondria are widely known to carry out oxidative phosphorylation. In addition, mitochondria are also involved in heme synthesis. Heme serves as a prosthetic group for several proteins that constitute the complexes of mitochondrial electron transport chain. Therefore, heme plays a pivotal role in OXPHOS and oxygen consumption. Further, lung cancer cells exhibit heme accumulation and require heme for proliferation and invasion in vitro. Abnormalities in mitochondrial biogenesis and mutations are implicated in cancer. This review delves into mitochondrial OXPHOS and lesser explored area of heme metabolism in lung cancer.
Collapse
Affiliation(s)
| | - Keely E FitzGerald
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX USA
| | | | - Chantal Vidal
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX USA
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX USA
| |
Collapse
|
38
|
Giedt RJ, Pathania D, Carlson JCT, McFarland PJ, Del Castillo AF, Juric D, Weissleder R. Single-cell barcode analysis provides a rapid readout of cellular signaling pathways in clinical specimens. Nat Commun 2018; 9:4550. [PMID: 30382095 PMCID: PMC6208406 DOI: 10.1038/s41467-018-07002-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/06/2018] [Indexed: 12/23/2022] Open
Abstract
Serial tissue sampling has become essential in guiding modern targeted and personalized cancer treatments. An alternative to image guided core biopsies are fine needle aspirates (FNA) that yield cells rather than tissues but are much better tolerated and have lower complication rates. The efficient pathway analysis of such cells in the clinic has been difficult, time consuming and costly. Here we develop an antibody-DNA barcoding approach where harvested cells can be rapidly re-stained through the use of custom designed oligonucleotide-fluorophore conjugates. We show that this approach can be used to interrogate drug-relevant pathways in scant clinical samples. Using the PI3K/PTEN/CDK4/6 pathways in breast cancer as an example, we demonstrate how analysis can be performed in tandem with trial enrollment and can evaluate downstream signaling following therapeutic inhibition. This approach should allow more widespread use of scant single cell material in clinical samples.
Collapse
Affiliation(s)
- Randy J Giedt
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Divya Pathania
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Jonathan C T Carlson
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
- MGH Cancer Center, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Philip J McFarland
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | | | - Dejan Juric
- MGH Cancer Center, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114, USA.
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
39
|
Harwig MC, Viana MP, Egner JM, Harwig JJ, Widlansky ME, Rafelski SM, Hill RB. Methods for imaging mammalian mitochondrial morphology: A prospective on MitoGraph. Anal Biochem 2018; 552:81-99. [PMID: 29505779 DOI: 10.1016/j.ab.2018.02.022] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/06/2018] [Accepted: 02/22/2018] [Indexed: 12/22/2022]
Abstract
Mitochondria are found in a variety of shapes, from small round punctate structures to a highly interconnected web. This morphological diversity is important for function, but complicates quantification. Consequently, early quantification efforts relied on various qualitative descriptors that understandably reduce the complexity of the network leading to challenges in consistency across the field. Recent application of state-of-the-art computational tools have resulted in more quantitative approaches. This prospective highlights the implementation of MitoGraph, an open-source image analysis platform for measuring mitochondrial morphology initially optimized for use with Saccharomyces cerevisiae. Here Mitograph was assessed on five different mammalian cells types, all of which were accurately segmented by MitoGraph analysis. MitoGraph also successfully differentiated between distinct mitochondrial morphologies that ranged from entirely fragmented to hyper-elongated. General recommendations are also provided for confocal imaging of labeled mitochondria (using mito-YFP, MitoTracker dyes and immunostaining parameters). Widespread adoption of MitoGraph will help achieve a long-sought goal of consistent and reproducible quantification of mitochondrial morphology.
Collapse
Affiliation(s)
- Megan C Harwig
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Matheus P Viana
- Visual Analytics and Comprehension Group, IBM Research, Brazil; Department of Developmental and Cell Biology and Center for Complex Biological Systems, University of California Irvine, Irvine, CA, 92697, United States
| | - John M Egner
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | | | - Michael E Widlansky
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Susanne M Rafelski
- Department of Developmental and Cell Biology and Center for Complex Biological Systems, University of California Irvine, Irvine, CA, 92697, United States; Assay Development, Allen Institute for Cell Science, Seattle, WA, 98109, United States
| | - R Blake Hill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, United States.
| |
Collapse
|
40
|
Abunimer AN, Mohammed H, Cook KL, Soto-Pantoja DR, Campos MM, Abu-Asab MS. Mitochondrial autophagosomes as a mechanism of drug resistance in breast carcinoma. Ultrastruct Pathol 2018; 42:170-180. [PMID: 29419344 PMCID: PMC6060621 DOI: 10.1080/01913123.2017.1419328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have previously described the process by which mitochondria donate their membranes for the formation of autophagosomes, and in this study we show that the same process could be involved in drug sequestration and exocytosis resulting in multidrug-resistant cancerous cells. We examine the implications of mitochondrial vesicle formation of mitoautophagosomes (MAPS) in response to the cytotoxic drug MKT-077, which targets mortalin, in a drug-resistant breast carcinoma cell line overexpressing P-glycoprotein (P-gp). The breast cancer cell line MCF-7Adr is derived from MCF-7, but differs from its ancestral line in tolerance of MKT-077-induced mitochondrial toxicity. Our ultrastructural observations suggest that autophagy in the MCF-7Adr cells entails regional sequestration of MKT077 in multilamellar LC3-labeled MAPS, which then separate from their mitochondria, and fuse with or engulf each other. MAPS appeared to be migrating through the cytoplasm and fusing with the plasma membrane, thus carrying out exocytotic secretion. This mechanism, which seems ineffective in the ancestral cell line, provides a resistance mechanism for MKT-077 by enhancing the efflux process of the cells. After 8 hr of MKT-077 exposure, a fraction of the resistant cells appeared viable and contained larger number of smaller sized mitochondria. Mitoautophagosomes, therefore, provide a potentially novel model for multidrug resistance in cancerous cells and may contribute to the P-gp efflux process.
Collapse
Affiliation(s)
- Ayman N. Abunimer
- Virginia Tech Carilion School of Medicine and Research Institute, Roanoke, VA, USA
| | - Heba Mohammed
- Section of Histopathology, National Eye Institute, NIH, Bethesda, MD, USA
| | - Katherine L. Cook
- Department of Surgery and Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - David R. Soto-Pantoja
- Department of Surgery and Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Mones S. Abu-Asab
- Section of Histopathology, National Eye Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
41
|
Pendin D, Filadi R, Pizzo P. The Concerted Action of Mitochondrial Dynamics and Positioning: New Characters in Cancer Onset and Progression. Front Oncol 2017; 7:102. [PMID: 28589083 PMCID: PMC5439081 DOI: 10.3389/fonc.2017.00102] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/02/2017] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are dynamic organelles whose morphology and activity are extremely variable, depending on the metabolic state of the cell. In particular, their shape and movements within the cell are finely regulated by an increasing number of proteins, which take part in the process of mitochondrial fission/fusion and connect the organelles to the cytoskeleton. As to their activities, mitochondria are considered to be at the crossroad between cell life and death since, on the one hand, they are essential in ATP production and in multiple metabolic pathways but, on the other, they are involved in the intrinsic apoptotic cascade, triggered by different stress conditions. Importantly, the process of mitochondrial Ca2+ uptake, as well as the morphology and the dynamics of these organelles, is known to deeply impact on both pro-survival and pro-death mitochondrial activities. Recently, increasing evidence has accrued on a central role of deregulated mitochondrial functionalities in the onset and progression of different pathologies, ranging from neurodegenerative diseases to cancer. In this contribution, we will present the latest findings connecting alterations in the machineries that control mitochondrial dynamics and localization to specific cancer hallmarks, highlighting the importance of mitochondria for the viability of cancer cells and discussing their role as promising targets for the development of novel anticancer therapies.
Collapse
Affiliation(s)
- Diana Pendin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Neuroscience Institute, National Research Council (CNR), Padova, Italy
| |
Collapse
|
42
|
Schcolnik-Cabrera A, Chávez-Blanco A, Domínguez-Gómez G, Dueñas-González A. Understanding tumor anabolism and patient catabolism in cancer-associated cachexia. Am J Cancer Res 2017; 7:1107-1135. [PMID: 28560061 PMCID: PMC5446478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/03/2017] [Indexed: 06/07/2023] Open
Abstract
Cachexia is a multifactorial paraneoplastic syndrome commonly associated with advanced stages of cancer. Cachexia is responsible for poor responses to antitumoral treatment and death in close to one-third of affected patients. There is still an incomplete understanding of the metabolic dysregulation induced by a tumor that leads to the appearance and persistence of cachexia. Furthermore, cachexia is irreversible, and there are currently no guidelines for its diagnosis or treatments for it. In this review, we aim to discuss the current knowledge about cancer-associated cachexia, starting with generalities about cancer as the generator of this syndrome, then analyzing the characteristics of cachexia at the biochemical and metabolic levels in both the tumor and the patient, and finally discussing current therapeutic approaches to treating cancer-associated cachexia.
Collapse
Affiliation(s)
| | | | | | - Alfonso Dueñas-González
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas UNAM/Instituto Nacional de CancerologíaMexico
| |
Collapse
|
43
|
Aouacheria A, Baghdiguian S, Lamb HM, Huska JD, Pineda FJ, Hardwick JM. Connecting mitochondrial dynamics and life-or-death events via Bcl-2 family proteins. Neurochem Int 2017; 109:141-161. [PMID: 28461171 DOI: 10.1016/j.neuint.2017.04.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/17/2017] [Indexed: 12/12/2022]
Abstract
The morphology of a population of mitochondria is the result of several interacting dynamical phenomena, including fission, fusion, movement, elimination and biogenesis. Each of these phenomena is controlled by underlying molecular machinery, and when defective can cause disease. New understanding of the relationships between form and function of mitochondria in health and disease is beginning to be unraveled on several fronts. Studies in mammals and model organisms have revealed that mitochondrial morphology, dynamics and function appear to be subject to regulation by the same proteins that regulate apoptotic cell death. One protein family that influences mitochondrial dynamics in both healthy and dying cells is the Bcl-2 protein family. Connecting mitochondrial dynamics with life-death pathway forks may arise from the intersection of Bcl-2 family proteins with the proteins and lipids that determine mitochondrial shape and function. Bcl-2 family proteins also have multifaceted influences on cells and mitochondria, including calcium handling, autophagy and energetics, as well as the subcellular localization of mitochondrial organelles to neuronal synapses. The remarkable range of physical or functional interactions by Bcl-2 family proteins is challenging to assimilate into a cohesive understanding. Most of their effects may be distinct from their direct roles in apoptotic cell death and are particularly apparent in the nervous system. Dual roles in mitochondrial dynamics and cell death extend beyond BCL-2 family proteins. In this review, we discuss many processes that govern mitochondrial structure and function in health and disease, and how Bcl-2 family proteins integrate into some of these processes.
Collapse
Affiliation(s)
- Abdel Aouacheria
- Institute of Evolutionary Sciences of Montpellier (ISEM), CNRS UMR 5554, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Stephen Baghdiguian
- Institute of Evolutionary Sciences of Montpellier (ISEM), CNRS UMR 5554, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Heather M Lamb
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - Jason D Huska
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - Fernando J Pineda
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA; Department of Biostatistics, Johns Hopkins University, Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA.
| |
Collapse
|
44
|
Thomas LW, Staples O, Turmaine M, Ashcroft M. CHCHD4 Regulates Intracellular Oxygenation and Perinuclear Distribution of Mitochondria. Front Oncol 2017; 7:71. [PMID: 28497026 PMCID: PMC5406405 DOI: 10.3389/fonc.2017.00071] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/28/2017] [Indexed: 11/25/2022] Open
Abstract
Hypoxia is a characteristic of the tumor microenvironment and is known to contribute to tumor progression and treatment resistance. Hypoxia-inducible factor (HIF) dimeric transcription factors control the cellular response to reduced oxygenation by regulating the expression of genes involved in metabolic adaptation, cell motility, and survival. Alterations in mitochondrial metabolism are not only a downstream consequence of HIF-signaling but mitochondria reciprocally regulate HIF signaling through multiple means, including oxygen consumption, metabolic intermediates, and reactive oxygen species generation. CHCHD4 is a redox-sensitive mitochondrial protein, which we previously identified and showed to be a novel regulator of HIF and hypoxia responses in tumors. Elevated expression of CHCHD4 in human tumors correlates with the hypoxia gene signature, disease progression, and poor patient survival. Here, we show that either long-term (72 h) exposure to hypoxia (1% O2) or elevated expression of CHCHD4 in tumor cells in normoxia leads to perinuclear accumulation of mitochondria, which is dependent on the expression of HIF-1α. Furthermore, we show that CHCHD4 is required for perinuclear localization of mitochondria and HIF activation in response to long-term hypoxia. Mutation of the functionally important highly conserved cysteines within the Cys-Pro-Cys motif of CHCHD4 or inhibition of complex IV activity (by sodium azide) redistributes mitochondria from the perinuclear region toward the periphery of the cell and blocks HIF activation. Finally, we show that CHCHD4-mediated perinuclear localization of mitochondria is associated with increased intracellular hypoxia within the perinuclear region and constitutive basal HIF activation in normoxia. Our study demonstrates that the intracellular distribution of the mitochondrial network is an important feature of the cellular response to hypoxia, contributing to hypoxic signaling via HIF activation and regulated by way of the cross talk between CHCHD4 and HIF-1α.
Collapse
Affiliation(s)
- Luke W. Thomas
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Oliver Staples
- Centre for Cell Signalling and Molecular Genetics, Division of Medicine, University College London, London, UK
| | - Mark Turmaine
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Margaret Ashcroft
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|