101
|
Guha S, Cheng A, Carroll T, King D, Koren SA, Swords S, Nehrke K, Johnson GVW. Selective disruption of Drp1-independent mitophagy and mitolysosome trafficking by an Alzheimer's disease relevant tau modification in a novel Caenorhabditis elegans model. Genetics 2022; 222:iyac104. [PMID: 35916724 PMCID: PMC9434186 DOI: 10.1093/genetics/iyac104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/06/2022] [Indexed: 11/14/2022] Open
Abstract
Accumulation of inappropriately phosphorylated tau into neurofibrillary tangles is a defining feature of Alzheimer's disease, with Tau pT231 being an early harbinger of tau pathology. Previously, we demonstrated that expressing a single genomic copy of human phosphomimetic mutant tau (T231E) in Caenorhabditis elegans drove age-dependent neurodegeneration. A critical finding was that T231E, unlike wild-type tau, completely and selectively suppressed oxidative stress-induced mitophagy. Here, we used dynamic imaging approaches to analyze T231E-associated changes in mitochondria and mitolysosome morphology, abundance, trafficking, and stress-induced mitophagy as a function of mitochondrial fission mediator dynamin-related protein 1, which has been demonstrated to interact with hyper phosphorylated tau and contribute to Alzheimer's disease pathogenesis, as well as Pink1, a well-recognized mediator of mitochondrial quality control that works together with Parkin to support stress-induced mitophagy. T231E impacted both mitophagy and mitolysosome neurite trafficking with exquisite selectivity, sparing macroautophagy as well as lysosome and autolysosome trafficking. Both oxidative-stress-induced mitophagy and the ability of T231E to suppress it were independent of drp-1, but at least partially dependent on pink-1. Organelle trafficking was more complicated, with drp-1 and pink-1 mutants exerting independent effects, but generally supported the idea that the mitophagy phenotype is of greater physiologic impact in T231E. Collectively, our results refine the mechanistic pathway through which T231E causes neurodegeneration, demonstrating pathologic selectivity for mutations that mimic tauopathy-associated post-translational modifications, physiologic selectivity for organelles that contain damaged mitochondria, and molecular selectivity for dynamin-related protein 1-independent, Pink1-dependent, perhaps adaptive, and mitophagy.
Collapse
Affiliation(s)
- Sanjib Guha
- Department of Anesthesiology & Perioperative Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Anson Cheng
- Department of Anesthesiology & Perioperative Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Trae Carroll
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Dennisha King
- Department of Neuroscience, University of Rochester, Rochester, NY 14642, USA
| | - Shon A Koren
- Department of Anesthesiology & Perioperative Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Sierra Swords
- Department of Molecular Biology and Biochemistry, Rutgers University, New Brunswick, NJ 08901, USA
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester, Rochester, NY 14642, USA
| | - Gail V W Johnson
- Department of Anesthesiology & Perioperative Medicine, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
102
|
Wang C, Fu H, Tan J, Zhang X. Rational Design of Oxazolidine-Based Red Fluorescent pH Probe for Simultaneous Imaging Two Subcellular Organelles. BIOSENSORS 2022; 12:bios12090696. [PMID: 36140081 PMCID: PMC9496385 DOI: 10.3390/bios12090696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022]
Abstract
A reversible pH-responsive fluorescent probe, BP, was rationally designed and synthesized, based on protonation and deprotonation gave rise to oxazolidine ring open and close. The fluorescence response of BP against pH ranges from 3.78 to 7.54, which is suitable for labeling intracellular pH-dependent organelles. BP displayed strong red emission at a relatively high pH in living HeLa cells and U87 cells. More importantly, this probe exhibited good colocalization with both mitochondria and lysosomes in these two cell lines, attributing to pH-induced structure tautomerism resulting in an oxazolidine ring open and close that triggered effective targeting of these two organelles. As organelle interactions are critical for cellular processes, this strategy of targeting dual organelles through the structure tautomerism is conducive to further developing more effective and advanced probes for real-time imaging of the interaction between mitochondria and lysosomes.
Collapse
Affiliation(s)
- Chunfei Wang
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Hengyi Fu
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Jingyun Tan
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Xuanjun Zhang
- Faculty of Health Sciences, University of Macau, Macau 999078, China
- MOE Frontiers Science Centre for Precision Oncology, University of Macau, Macau 999078, China
- Correspondence:
| |
Collapse
|
103
|
Lippi A, Krisko A. CORE at the boundary of stress resistance and longevity. Int J Biochem Cell Biol 2022; 151:106277. [PMID: 35995386 DOI: 10.1016/j.biocel.2022.106277] [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: 06/09/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 10/15/2022]
Abstract
As chronological age of an organism increases, a number of errors accumulate at different levels of biological organization. The tendency of errors to accumulate and cause downstream problems in maintenance of cellular homeostasis is met by numerous protection and repair mechanisms. Maintenance of proteins is vital for cell viability and longevity, thus cellular proteostasis is supported by chaperone networks in every cellular compartment, as well as other pathways ensuring timely chaperone expression and activity. In this minireview, we summarize the progress related to the cross-organelle stress response (CORE), in charge of orchestrating a cell-wide response to compartmentalized proteotoxicity. The proposed CORE pathway encompasses activation of protein conformational maintenance machineries, antioxidant enzymes and metabolic changes simultaneously in the cytosol, mitochondria and the ER. We discuss its importance in cell survival and longevity as well as its potential to serve as a pharmaceutical target in age-related diseases.
Collapse
Affiliation(s)
- Alice Lippi
- Department of Experimental Neurodegeneration, University Medical Center Goettingen, Waldweg 33, 37075 Goettingen, Germany
| | - Anita Krisko
- Department of Experimental Neurodegeneration, University Medical Center Goettingen, Waldweg 33, 37075 Goettingen, Germany.
| |
Collapse
|
104
|
Yang Q, Fang Y, Zhang C, Liu X, Wu Y, Zhang Y, Yang J, Yong K. Exposure to zinc induces lysosomal-mitochondrial axis-mediated apoptosis in PK-15 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113716. [PMID: 35667309 DOI: 10.1016/j.ecoenv.2022.113716] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
Zinc (Zn), a kind of metallic element, can cause poisonous effects on host physiology when its excess exposure. Lysosomes and mitochondria are the toxic targets of heavy metals, and the lysosomal-mitochondrial axis is also verified to take part in apoptosis, but the related underlying mechanisms in Zn-induced cytotoxicity remain undefined. Here, we identified that excess Zn could cause cell damage in PK-15 cells accompanied by the lysosomal and mitochondrial dysfunction, with the evidence by the elevated levels of cathepsin B/D (CTSB/CTSD) in cytoplasm and decrease of Lyso-Tracker Red signal, red fluorescence intensity of AO staining, mitochondrial complex enzyme activities and ATP production. Additionally, the number of Annexin V+/PI--stained cells, apoptosis-related genes (Bax, Bid, Bak1, Caspase-9, and Caspase-3) and proteins levels of Bax, Bak1, Caspase-9, cleaved Caspase-3 and cytoplasmic Cyt C were signally elevated under Zn exposure, while the protein levels of Bcl2 and mitochondrial Cyt C were observably decreased. Importantly, Pepstatin A (the activity inhibitor of CTSD) and RNA interference of CTSD (si-CTSD) was used to reduce the release of lysosomal CTSD to the cytoplasm, which could signally alleviated Zn-induced mitochondrial damage and apoptosis. In summary, these results suggested that Zn could induced lysosomal and mitochondrial dysfunction in PK-15 cells, and the CTSD played an important role in Zn-induced lysosomal-mitochondrial axis-mediated apoptosis. Our results provided a new insight in Zn-induced toxicology, which for protecting the ecological environment and public health.
Collapse
Affiliation(s)
- Qingwen Yang
- Laboratory of Veterinary Pharmacology, Department of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, PR China
| | - Yudong Fang
- College of Life Sciences, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, PR China
| | - Chuanshi Zhang
- Laboratory of Veterinary Pharmacology, Department of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, PR China
| | - Xuesong Liu
- Laboratory of Veterinary Pharmacology, Department of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, PR China
| | - Youhua Wu
- Laboratory of Veterinary Pharmacology, Department of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, PR China
| | - Yi Zhang
- Laboratory of Veterinary Pharmacology, Department of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, PR China
| | - Junjie Yang
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, PR China
| | - Kang Yong
- Laboratory of Veterinary Pharmacology, Department of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, PR China.
| |
Collapse
|
105
|
Gu Y, Guerra F, Hu M, Pope A, Sung K, Yang W, Jetha S, Shoff TA, Gunatilake T, Dahlkamp O, Shi LZ, Manganelli F, Nolano M, Zhou Y, Ding J, Bucci C, Wu C. Mitochondria dysfunction in Charcot Marie Tooth 2B Peripheral Sensory Neuropathy. Commun Biol 2022; 5:717. [PMID: 35851620 PMCID: PMC9293960 DOI: 10.1038/s42003-022-03632-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 06/23/2022] [Indexed: 11/21/2022] Open
Abstract
Rab7 GTPase regulates mitochondrial morphology and function. Missense mutation(s) of Rab7 underlies the pathogenesis of Charcot Marie Tooth 2B (CMT2B) peripheral neuropathy. Herein, we investigate how mitochondrial morphology and function are impacted by the CMT2B associated Rab7V162M mutation. In contrast to recent studies of using heterologous overexpression systems, our results demonstrate significant mitochondrial fragmentation in both human CMT2B patient fibroblasts and CMT2B embryonic fibroblasts (MEFs). Primary cultured E18 dorsal root ganglion (DRG) sensory neurons also show mitochondrial fragmentation and altered axonal mitochondrial movement. In addition, we demonstrate that inhibitors to either the mitochondrial fission protein Drp1 or to the nucleotide binding to Rab7 normalize the mitochondrial deficits in both MEFs and E18 cultured DRG neurons. Our study reveals, for the first time, that expression of CMT2B Rab7 mutation at the physiological level enhances Drp1 activity to promote mitochondrial fission, potentially underlying selective vulnerability of peripheral sensory neurons in CMT2B pathogenesis.
Collapse
Affiliation(s)
- Yingli Gu
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
- Department of Neurology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), Università del Salento, Via Provinciale Lecce-Monteroni n. 165, 73100, Lecce, Italy
| | - Mingzheng Hu
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Alexander Pope
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Kijung Sung
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Wanlin Yang
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
- Department of Neurology, Zhujiang Hospital of Southern Medical University Guangzhou, Guangzhou, 510280, Guangdong Sheng, China
| | - Simone Jetha
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Thomas A Shoff
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Tessanya Gunatilake
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Owen Dahlkamp
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Linda Zhixia Shi
- Department of Bioengineering, University of California San Diego, La Jolla, 92093, CA, USA
| | - Fiore Manganelli
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Via Sergio Pansini 5, 80131, Naples, Italy
| | - Maria Nolano
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Via Sergio Pansini 5, 80131, Naples, Italy
| | - Yue Zhou
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030, China
| | - Jianqing Ding
- Institute of Neurology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), Università del Salento, Via Provinciale Lecce-Monteroni n. 165, 73100, Lecce, Italy.
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA.
| |
Collapse
|
106
|
Lang M, Pramstaller PP, Pichler I. Crosstalk of organelles in Parkinson's disease - MiT family transcription factors as central players in signaling pathways connecting mitochondria and lysosomes. Mol Neurodegener 2022; 17:50. [PMID: 35842725 PMCID: PMC9288732 DOI: 10.1186/s13024-022-00555-7] [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: 11/10/2021] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
Living organisms constantly need to adapt to their surrounding environment and have evolved sophisticated mechanisms to deal with stress. Mitochondria and lysosomes are central organelles in the response to energy and nutrient availability within a cell and act through interconnected mechanisms. However, when such processes become overwhelmed, it can lead to pathologies. Parkinson's disease (PD) is a common neurodegenerative disorder (NDD) characterized by proteinaceous intracellular inclusions and progressive loss of dopaminergic neurons, which causes motor and non-motor symptoms. Genetic and environmental factors may contribute to the disease etiology. Mitochondrial dysfunction has long been recognized as a hallmark of PD pathogenesis, and several aspects of mitochondrial biology are impaired in PD patients and models. In addition, defects of the autophagy-lysosomal pathway have extensively been observed in cell and animal models as well as PD patients' brains, where constitutive autophagy is indispensable for adaptation to stress and energy deficiency. Genetic and molecular studies have shown that the functions of mitochondria and lysosomal compartments are tightly linked and influence each other. Connections between these organelles are constituted among others by mitophagy, organellar dynamics and cellular signaling cascades, such as calcium (Ca2+) and mTOR (mammalian target of rapamycin) signaling and the activation of transcription factors. Members of the Microphthalmia-associated transcription factor family (MiT), including MITF, TFE3 and TFEB, play a central role in regulating cellular homeostasis in response to metabolic pressure and are considered master regulators of lysosomal biogenesis. As such, they are part of the interconnection between mitochondria and lysosome functions and therefore represent attractive targets for therapeutic approaches against NDD, including PD. The activation of MiT transcription factors through genetic and pharmacological approaches have shown encouraging results at ameliorating PD-related phenotypes in in vitro and in vivo models. In this review, we summarize the relationship between mitochondrial and autophagy-lysosomal functions in the context of PD etiology and focus on the role of the MiT pathway and its potential as pharmacological target against PD.
Collapse
Affiliation(s)
- Martin Lang
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.,Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| |
Collapse
|
107
|
Han JH, Kim YK, Kim H, Lee J, Oh MJ, Kim SB, Kim M, Kim KH, Yoon HJ, Lee MS, Minna JD, White MA, Kim HS. Snail acetylation by autophagy-derived acetyl-coenzyme A promotes invasion and metastasis of KRAS-LKB1 co-mutated lung cancer cells. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:716-749. [PMID: 35838183 PMCID: PMC9395322 DOI: 10.1002/cac2.12332] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/21/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
Background Autophagy is elevated in metastatic tumors and is often associated with active epithelial‐to‐mesenchymal transition (EMT). However, the extent to which EMT is dependent on autophagy is largely unknown. This study aimed to identify the mechanisms by which autophagy facilitates EMT. Methods We employed a liquid chromatography‐based metabolomic approach with kirsten rat sarcoma viral oncogene (KRAS) and liver kinase B1 (LKB1) gene co‐mutated (KL) cells that represent an autophagy/EMT‐coactivated invasive lung cancer subtype for the identification of metabolites linked to autophagy‐driven EMT activation. Molecular mechanisms of autophagy‐driven EMT activation were further investigated by quantitative real‐time polymerase chain reaction (qRT‐PCR), Western blotting analysis, immunoprecipitation, immunofluorescence staining, and metabolite assays. The effects of chemical and genetic perturbations on autophagic flux were assessed by two orthogonal approaches: microtubule‐associated protein 1A/1B‐light chain 3 (LC3) turnover analysis by Western blotting and monomeric red fluorescent protein‐green fluorescent protein (mRFP‐GFP)‐LC3 tandem fluorescent protein quenching assay. Transcription factor EB (TFEB) activity was measured by coordinated lysosomal expression and regulation (CLEAR) motif‐driven luciferase reporter assay. Experimental metastasis (tail vein injection) mouse models were used to evaluate the impact of calcium/calmodulin‐dependent protein kinase kinase 2 (CAMKK2) or ATP citrate lyase (ACLY) inhibitors on lung metastasis using IVIS luciferase imaging system. Results We found that autophagy in KL cancer cells increased acetyl‐coenzyme A (acetyl‐CoA), which facilitated the acetylation and stabilization of the EMT‐inducing transcription factor Snail. The autophagy/acetyl‐CoA/acetyl‐Snail axis was further validated in tumor tissues and in autophagy‐activated pancreatic cancer cells. TFEB acetylation in KL cancer cells sustained pro‐metastatic autophagy in a mammalian target of rapamycin complex 1 (mTORC1)‐independent manner. Pharmacological inhibition of this axis via CAMKK2 inhibitors or ACLY inhibitors consistently reduced the metastatic capacity of KL cancer cells in vivo. Conclusions This study demonstrates that autophagy‐derived acetyl‐CoA promotes Snail acetylation and thereby facilitates invasion and metastasis of KRAS‐LKB1 co‐mutated lung cancer cells and that inhibition of the autophagy/acetyl‐CoA/acetyl‐Snail axis using CAMKK2 or ACLY inhibitors could be a potential therapeutic strategy to suppress metastasis of KL lung cancer.
Collapse
Affiliation(s)
- Jang Hee Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Department of Medical Science, Yonsei University Graduate School, Seoul, 03722, Korea.,Department of Urology, Seoul National University Hospital, Seoul, 03722, Korea
| | - Yong Keon Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hakhyun Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jooyoung Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Checkmate Therapeutics Inc., Seoul, 07207, Korea
| | - Myung Joon Oh
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Sang Bum Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Minjee Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kook Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hyun Ju Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Myung-Shik Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Michael A White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Hyun Seok Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Checkmate Therapeutics Inc., Seoul, 07207, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| |
Collapse
|
108
|
Barzegari A, Aaboulhassanzadeh S, Landon R, Gueguen V, Meddahi-Pellé A, Parvizpour S, Anagnostou F, Pavon-Djavid G. Mitohormesis and mitochondrial dynamics in the regulation of stem cell fate. J Cell Physiol 2022; 237:3435-3448. [PMID: 35775725 DOI: 10.1002/jcp.30820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/11/2022]
Abstract
The ability of stem cells for self-renewing, differentiation, and regeneration of injured tissues is believed to occur via the hormetic modulation of nuclear/mitochondrial signal transductions. The evidence now indicates that in damaged tissues, the mitochondria set off the alarm under oxidative stress conditions, hence they are the central regulators of stem cell fate decisions. This review aimed to provide an update to a broader concept of stem cell fate in stress conditions of damaged tissues, and insights for the mitochondrial hormesis (mitohormesis), including the integrated stress response (ISR), mitochondrial dynamics, mitochondria uncoupling, unfolded protein response, and mitokines, with implications for the control of stem cells programing in a successful clinical cell therapy.
Collapse
Affiliation(s)
- Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sobhan Aaboulhassanzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rebecca Landon
- CNRS UMR7052-INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Paris Diderot University, Paris, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Sepideh Parvizpour
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fani Anagnostou
- CNRS UMR7052-INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Paris Diderot University, Paris, France
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| |
Collapse
|
109
|
Remission of obesity and insulin resistance is not sufficient to restore mitochondrial homeostasis in visceral adipose tissue. Redox Biol 2022; 54:102353. [PMID: 35777200 PMCID: PMC9287736 DOI: 10.1016/j.redox.2022.102353] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
Metabolic plasticity is the ability of a biological system to adapt its metabolic phenotype to different environmental stressors. We used a whole-body and tissue-specific phenotypic, functional, proteomic, metabolomic and transcriptomic approach to systematically assess metabolic plasticity in diet-induced obese mice after a combined nutritional and exercise intervention. Although most obesity and overnutrition-related pathological features were successfully reverted, we observed a high degree of metabolic dysfunction in visceral white adipose tissue, characterized by abnormal mitochondrial morphology and functionality. Despite two sequential therapeutic interventions and an apparent global healthy phenotype, obesity triggered a cascade of events in visceral adipose tissue progressing from mitochondrial metabolic and proteostatic alterations to widespread cellular stress, which compromises its biosynthetic and recycling capacity. In humans, weight loss after bariatric surgery showed a transcriptional signature in visceral adipose tissue similar to our mouse model of obesity reversion. Overall, our data indicate that obesity prompts a lasting metabolic fingerprint that leads to a progressive breakdown of metabolic plasticity in visceral adipose tissue.
Collapse
|
110
|
Vrijsen S, Vrancx C, Del Vecchio M, Swinnen JV, Agostinis P, Winderickx J, Vangheluwe P, Annaert W. Inter-organellar Communication in Parkinson's and Alzheimer's Disease: Looking Beyond Endoplasmic Reticulum-Mitochondria Contact Sites. Front Neurosci 2022; 16:900338. [PMID: 35801175 PMCID: PMC9253489 DOI: 10.3389/fnins.2022.900338] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/05/2022] [Indexed: 01/13/2023] Open
Abstract
Neurodegenerative diseases (NDs) are generally considered proteinopathies but whereas this may initiate disease in familial cases, onset in sporadic diseases may originate from a gradually disrupted organellar homeostasis. Herein, endolysosomal abnormalities, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and altered lipid metabolism are commonly observed in early preclinical stages of major NDs, including Parkinson's disease (PD) and Alzheimer's disease (AD). Among the multitude of underlying defective molecular mechanisms that have been suggested in the past decades, dysregulation of inter-organellar communication through the so-called membrane contact sites (MCSs) is becoming increasingly apparent. Although MCSs exist between almost every other type of subcellular organelle, to date, most focus has been put on defective communication between the ER and mitochondria in NDs, given these compartments are critical in neuronal survival. Contributions of other MCSs, notably those with endolysosomes and lipid droplets are emerging, supported as well by genetic studies, identifying genes functionally involved in lysosomal homeostasis. In this review, we summarize the molecular identity of the organelle interactome in yeast and mammalian cells, and critically evaluate the evidence supporting the contribution of disturbed MCSs to the general disrupted inter-organellar homeostasis in NDs, taking PD and AD as major examples.
Collapse
Affiliation(s)
- Stephanie Vrijsen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, Leuven, Belgium
| | - Céline Vrancx
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Mara Del Vecchio
- Laboratory of Functional Biology, Department of Biology, KU Leuven, Heverlee, Belgium
| | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, VIB-Center for Cancer Research, KU Leuven, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joris Winderickx
- Laboratory of Functional Biology, Department of Biology, KU Leuven, Heverlee, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, Leuven, Belgium
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
111
|
Coelho P, Fão L, Mota S, Rego AC. Mitochondrial function and dynamics in neural stem cells and neurogenesis: Implications for neurodegenerative diseases. Ageing Res Rev 2022; 80:101667. [PMID: 35714855 DOI: 10.1016/j.arr.2022.101667] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/21/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
Mitochondria have been largely described as the powerhouse of the cell and recent findings demonstrate that this organelle is fundamental for neurogenesis. The mechanisms underlying neural stem cells (NSCs) maintenance and differentiation are highly regulated by both intrinsic and extrinsic factors. Mitochondrial-mediated switch from glycolysis to oxidative phosphorylation, accompanied by mitochondrial remodeling and dynamics are vital to NSCs fate. Deregulation of mitochondrial proteins, mitochondrial DNA, function, fission/fusion and metabolism underly several neurodegenerative diseases; data show that these impairments are already present in early developmental stages and NSC fate decisions. However, little is known about mitochondrial role in neurogenesis. In this Review, we describe the recent evidence covering mitochondrial role in neurogenesis, its impact in selected neurodegenerative diseases, for which aging is the major risk factor, and the recent advances in stem cell-based therapies that may alleviate neurodegenerative disorders-related neuronal deregulation through improvement of mitochondrial function and dynamics.
Collapse
Affiliation(s)
- Patrícia Coelho
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal.
| | - Lígia Fão
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| | - Sandra Mota
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; III, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - A Cristina Rego
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| |
Collapse
|
112
|
Zhang J, Shi Y. In Search of the Holy Grail: Toward a Unified Hypothesis on Mitochondrial Dysfunction in Age-Related Diseases. Cells 2022; 11:cells11121906. [PMID: 35741033 PMCID: PMC9221202 DOI: 10.3390/cells11121906] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiolipin (CL) is a mitochondrial signature phospholipid that plays a pivotal role in mitochondrial dynamics, membrane structure, oxidative phosphorylation, mtDNA bioenergetics, and mitophagy. The depletion or abnormal acyl composition of CL causes mitochondrial dysfunction, which is implicated in the pathogenesis of aging and age-related disorders. However, the molecular mechanisms by which mitochondrial dysfunction causes age-related diseases remain poorly understood. Recent development in the field has identified acyl-CoA:lysocardiolipin acyltransferase 1 (ALCAT1), an acyltransferase upregulated by oxidative stress, as a key enzyme that promotes mitochondrial dysfunction in age-related diseases. ALCAT1 catalyzes CL remodeling with very-long-chain polyunsaturated fatty acids, such as docosahexaenoic acid (DHA). Enrichment of DHA renders CL highly sensitive to oxidative damage by reactive oxygen species (ROS). Oxidized CL becomes a new source of ROS in the form of lipid peroxides, leading to a vicious cycle of oxidative stress, CL depletion, and mitochondrial dysfunction. Consequently, ablation or the pharmacological inhibition of ALCAT1 have been shown to mitigate obesity, type 2 diabetes, heart failure, cardiomyopathy, fatty liver diseases, neurodegenerative diseases, and cancer. The findings suggest that age-related disorders are one disease (aging) manifested by different mitochondrion-sensitive tissues, and therefore should be treated as one disease. This review will discuss a unified hypothesis on CL remodeling by ALCAT1 as the common denominator of mitochondrial dysfunction, linking mitochondrial dysfunction to the development of age-related diseases.
Collapse
Affiliation(s)
| | - Yuguang Shi
- Correspondence: ; Tel.: +1-210-450-1363; Fax: +1-210-562-6150
| |
Collapse
|
113
|
Jain A, Zoncu R. Organelle transporters and inter-organelle communication as drivers of metabolic regulation and cellular homeostasis. Mol Metab 2022; 60:101481. [PMID: 35342037 PMCID: PMC9043965 DOI: 10.1016/j.molmet.2022.101481] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Spatial compartmentalization of metabolic pathways within membrane-separated organelles is key to the ability of eukaryotic cells to precisely regulate their biochemical functions. Membrane-bound organelles such as mitochondria, endoplasmic reticulum (ER) and lysosomes enable the concentration of metabolic precursors within optimized chemical environments, greatly accelerating the efficiency of both anabolic and catabolic reactions, enabling division of labor and optimal utilization of resources. However, metabolic compartmentalization also poses a challenge to cells because it creates spatial discontinuities that must be bridged for reaction cascades to be connected and completed. To do so, cells employ different methods to coordinate metabolic fluxes occurring in different organelles, such as membrane-localized transporters to facilitate regulated metabolite exchange between mitochondria and lysosomes, non-vesicular transport pathways via physical contact sites connecting the ER with both mitochondria and lysosomes, as well as localized regulatory signaling processes that coordinately regulate the activity of all these organelles. SCOPE OF REVIEW This review covers how cells use membrane transporters, membrane contact sites, and localized signaling pathways to mediate inter-organelle communication and coordinate metabolism. We also describe how disruption of inter-organelle communication is an emerging driver in a multitude of diseases, from cancer to neurodegeneration. MAJOR CONCLUSIONS Effective communication among organelles is essential to cellular health and function. Identifying the major molecular players involved in mediating metabolic coordination between organelles will further our understanding of cellular metabolism in health and lead us to design better therapeutics against dysregulated metabolism in disease.
Collapse
Affiliation(s)
- Aakriti Jain
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
114
|
Guo J, Liu K, Wang J, Jiang H, Zhang M, Liu Y, Shan C, Hu F, Fu W, Zhang C, Li J, Chen Y. A rational foundation for micheliolide-based combination strategy by targeting redox and metabolic circuit in cancer cells. Biochem Pharmacol 2022; 200:115037. [DOI: 10.1016/j.bcp.2022.115037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/24/2022] [Accepted: 04/06/2022] [Indexed: 11/02/2022]
|
115
|
Jethava KP, Prakash P, Manchanda P, Arora H, Chopra G. One Scaffold, Different Organelle Sensors: pH-Activable Fluorescent Probes for Targeting Live Microglial Cell Organelles. Chembiochem 2022; 23:e202100378. [PMID: 34585478 PMCID: PMC9835645 DOI: 10.1002/cbic.202100378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/28/2021] [Indexed: 01/14/2023]
Abstract
Targeting live cell organelles is essential for imaging, understanding, and controlling specific biochemical processes. Typically, fluorescent probes with distinct structural scaffolds are used to target specific cell organelles. Here, we have designed a modular one-step synthetic strategy using a common reaction intermediate to develop new lysosomal, mitochondrial, and nucleus-targeting pH-activable fluorescent probes that are all based on a single boron dipyrromethane scaffold. The divergent cell organelle targeting was achieved by synthesizing probes with specific functional group changes to the central scaffold resulting in differential fluorescence and pKa . Specifically, we show that the functional group transformation of the same scaffold influences cellular localization and specificity of pH-activable fluorescent probes in live primary microglial cells with pKa values ranging from ∼3.2-6.0. We introduce a structure-organelle-relationship (SOR) framework to target nuclei (NucShine), lysosomes (LysoShine), and mitochondria (MitoShine) in live microglia. This work will result in future applications of SOR beyond imaging to target and control organelle-specific biochemical processes in disease-specific models.
Collapse
Affiliation(s)
- Krupal P. Jethava
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 (USA)
| | - Priya Prakash
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 (USA)
| | - Palak Manchanda
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 (USA)
| | - Harshit Arora
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 (USA)
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 (USA),Purdue University, Purdue Institute for Drug Discovery, West Lafayette, IN 47907 (USA),Purdue University, Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907 (USA),Purdue University, Purdue Institute for Inflammation, Immunology and Infectious Disease, West Lafayette, IN 47907 (USA),Purdue University, Purdue Center for Cancer Research, West Lafayette, IN 47907 (USA),Purdue University, Integrative Data Science Initiative, West Lafayette, IN 47907 (USA)
| |
Collapse
|
116
|
Su X, Zhou M, Li Y, Zhang J, An N, Yang F, Zhang G, Yuan C, Chen H, Wu H, Xing Y. Protective effects of natural products against myocardial ischemia/reperfusion: Mitochondria-targeted therapeutics. Biomed Pharmacother 2022; 149:112893. [PMID: 35366532 DOI: 10.1016/j.biopha.2022.112893] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Patients with ischemic heart disease receiving reperfusion therapy still need to face left ventricular remodeling and heart failure after myocardial infarction. Reperfusion itself paradoxically leads to further cardiomyocyte death and systolic dysfunction. Ischemia/reperfusion (I/R) injury can eliminate the benefits of reperfusion therapy in patients and causes secondary myocardial injury. Mitochondrial dysfunction and structural disorder are the basic driving force of I/R injury. We summarized the basic relationship and potential mechanisms of mitochondrial injury in the development of I/R injury. Subsequently, this review summarized the natural products (NPs) that have been proven to targeting mitochondrial therapeutic effects during I/R injury in recent years and related cellular signal transduction pathways. We found that these NPs mainly protected the structural integrity of mitochondria and improve dysfunction, such as reducing mitochondrial division and fusion abnormalities, improving mitochondrial Ca2+ overload and inhibiting reactive oxygen species overproduction, thereby playing a role in protecting cardiomyocytes during I/R injury. This data would deepen the understanding of I/R-induced mitochondrial pathological process and suggested that NPs are expected to be transformed into potential therapies targeting mitochondria.
Collapse
Affiliation(s)
- Xin Su
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Mingyang Zhou
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yingjian Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jianzhen Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Na An
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoxia Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chao Yuan
- Dezhou Second People's Hospital, Dezhou 253000, China
| | - Hengwen Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Hongjin Wu
- Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital, Beijing 100191, China.
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
117
|
Zhang MY, Zhu L, Bao X, Xie TH, Cai J, Zou J, Wang W, Gu S, Li Y, Li HY, Yao Y, Wei TT. Inhibition of Drp1 ameliorates diabetic retinopathy by regulating mitochondrial homeostasis. Exp Eye Res 2022; 220:109095. [PMID: 35490835 DOI: 10.1016/j.exer.2022.109095] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/06/2022] [Accepted: 04/25/2022] [Indexed: 12/19/2022]
Abstract
Diabetic retinopathy (DR) is a potentially blinding complication resulting from diabetes mellitus (DM). Retinal vascular endothelial cells (RMECs) dysfunction occupies an important position in the pathogenesis of DR, and mitochondrial disorders play a vital role in RMECs dysfunction. However, the detailed mechanisms underlying DR-induced mitochondrial disorders in RMECs remain elusive. In the present study, we used High glucose (HG)-induced RMECs in vitro and streptozotocin (STZ)-induced Sprague-Dawley rats in vivo to explore the related mechanisms. We found that HG-induced mitochondrial dysfunction via mitochondrial Dynamin-related protein 1(Drp1)-mediated mitochondrial fission. Drp1 inhibitor, Mdivi-1, rescued HG-induced mitochondrial dysfunction. Protein Kinase Cδ (PKCδ) could induce phosphorylation of Drp1, and we found that HG induced phosphorylation of PKCδ. PKCδ inhibitor (Go 6983) or PKCδ siRNA reversed HG-induced phosphorylation of Drp1 and further mitochondrial dysfunction. The above studies indicated that HG increases mitochondrial fission via promoting PKCδ/Drp1 signaling. Drp1 induces excessive mitochondrial fission and produces damaged mitochondrial, and mitophagy plays a key role in clearing damaged mitochondrial. Our study showed that HG suppressed mitophagy via inhibiting LC3B-II formation and p62 degradation. 3-MA (autophagy inhibitor) aggravated HG-induced RMECs damage, while rapamycin (autophagy agonist) rescued the above phenomenon. Further studies were identified that HG inhibited mitophagy by down-regulation of the PINK1/Parkin signaling pathway, and PINK1 siRNA aggravated HG-induced RMECs damage. Further in-depth study, we propose that Drp1 promotion of Hexokinase II (HK-II) separation from mitochondria, thus inhibiting HK-II-PINK1-mediated mitophagy. In vivo, we found that intraretinal microvascular abnormalities (IRMA), including retinal vascular leakage, acellular capillaries, and apoptosis were increased in STZ-induced DR rats, which were reversed by pretreatment with Mdivi-1 or Rapamycin. Altogether, our findings provide new insight into the mechanisms underlying the regulation of mitochondrial homeostasis and provide a potential treatment strategy for Diabetic retinopathy.
Collapse
Affiliation(s)
- Meng-Yuan Zhang
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Lingpeng Zhu
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Xun Bao
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Tian-Hua Xie
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Jiping Cai
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Jian Zou
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Wenjuan Wang
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Shun Gu
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Yan Li
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Hong-Ying Li
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China
| | - Yong Yao
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China; Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, PR China.
| | - Ting-Ting Wei
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China.
| |
Collapse
|
118
|
Mitochondria-lysosome contact site dynamics and misregulation in neurodegenerative diseases. Trends Neurosci 2022; 45:312-322. [PMID: 35249745 PMCID: PMC8930467 DOI: 10.1016/j.tins.2022.01.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 02/06/2023]
Abstract
Neurons rely heavily on properly regulated mitochondrial and lysosomal homeostasis, with multiple neurodegenerative diseases linked to dysfunction in these two organelles. Interestingly, mitochondria-lysosome membrane contact sites have been identified as a key pathway mediating their crosstalk in neurons. Recent studies have further elucidated the regulation of mitochondria-lysosome contact dynamics via distinct tethering/untethering protein machinery. Moreover, this pathway has been shown to have additional functions in regulating organelle network dynamics and metabolite transfer between lysosomes and mitochondria. In this review, we highlight recent advances in the field of mitochondria-lysosome contact sites and their misregulation across multiple neurodegenerative disorders, which further underscore a potential role for this pathway in neuronal homeostasis and disease.
Collapse
|
119
|
Martelli F, Hernandes NH, Zuo Z, Wang J, Wong CO, Karagas NE, Roessner U, Rupasinghe T, Robin C, Venkatachalam K, Perry T, Batterham P, Bellen HJ. Low doses of the organic insecticide spinosad trigger lysosomal defects, elevated ROS, lipid dysregulation, and neurodegeneration in flies. eLife 2022; 11:e73812. [PMID: 35191376 PMCID: PMC8863376 DOI: 10.7554/elife.73812] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
Large-scale insecticide application is a primary weapon in the control of insect pests in agriculture. However, a growing body of evidence indicates that it is contributing to the global decline in population sizes of many beneficial insect species. Spinosad emerged as an organic alternative to synthetic insecticides and is considered less harmful to beneficial insects, yet its mode of action remains unclear. Using Drosophila, we show that low doses of spinosad antagonize its neuronal target, the nicotinic acetylcholine receptor subunit alpha 6 (nAChRα6), reducing the cholinergic response. We show that the nAChRα6 receptors are transported to lysosomes that become enlarged and increase in number upon low doses of spinosad treatment. Lysosomal dysfunction is associated with mitochondrial stress and elevated levels of reactive oxygen species (ROS) in the central nervous system where nAChRα6 is broadly expressed. ROS disturb lipid storage in metabolic tissues in an nAChRα6-dependent manner. Spinosad toxicity is ameliorated with the antioxidant N-acetylcysteine amide. Chronic exposure of adult virgin females to low doses of spinosad leads to mitochondrial defects, severe neurodegeneration, and blindness. These deleterious effects of low-dose exposures warrant rigorous investigation of its impacts on beneficial insects.
Collapse
Affiliation(s)
- Felipe Martelli
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | | | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Julia Wang
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Ching-On Wong
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences CenterHoustonUnited States
| | - Nicholas E Karagas
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences CenterHoustonUnited States
| | - Ute Roessner
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | | | - Charles Robin
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences CenterHoustonUnited States
| | - Trent Perry
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | - Philip Batterham
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Neurological Research Institute, Texas Children HospitalHoustonUnited States
- Howard Hughes Medical Institute, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
120
|
Huang YC, Hsu SM, Shie FS, Shiao YJ, Chao LJ, Chen HW, Yao HH, Chien MA, Lin CC, Tsay HJ. Reduced mitochondria membrane potential and lysosomal acidification are associated with decreased oligomeric Aβ degradation induced by hyperglycemia: A study of mixed glia cultures. PLoS One 2022; 17:e0260966. [PMID: 35073330 PMCID: PMC8786178 DOI: 10.1371/journal.pone.0260966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/20/2021] [Indexed: 01/21/2023] Open
Abstract
Diabetes is a risk factor for Alzheimer’s disease (AD), a chronic neurodegenerative disease. We and others have shown prediabetes, including hyperglycemia and obesity induced by high fat and high sucrose diets, is associated with exacerbated amyloid beta (Aβ) accumulation and cognitive impairment in AD transgenic mice. However, whether hyperglycemia reduce glial clearance of oligomeric amyloid-β (oAβ), the most neurotoxic Aβ aggregate, remains unclear. Mixed glial cultures simulating the coexistence of astrocytes and microglia in the neural microenvironment were established to investigate glial clearance of oAβ under normoglycemia and chronic hyperglycemia. Ramified microglia and low IL-1β release were observed in mixed glia cultures. In contrast, amoeboid-like microglia and higher IL-1β release were observed in primary microglia cultures. APPswe/PS1dE9 transgenic mice are a commonly used AD mouse model. Microglia close to senile plaques in APPswe/PS1dE9 transgenic mice exposed to normoglycemia or chronic hyperglycemia exhibited an amoeboid-like morphology; other microglia were ramified. Therefore, mixed glia cultures reproduce the in vivo ramified microglial morphology. To investigate the impact of sustained high-glucose conditions on glial oAβ clearance, mixed glia were cultured in media containing 5.5 mM glucose (normal glucose, NG) or 25 mM glucose (high glucose, HG) for 16 days. Compared to NG, HG reduced the steady-state level of oAβ puncta internalized by microglia and astrocytes and decreased oAβ degradation kinetics. Furthermore, the lysosomal acidification and lysosomal hydrolysis activity of microglia and astrocytes were lower in HG with and without oAβ treatment than NG. Moreover, HG reduced mitochondrial membrane potential and ATP levels in mixed glia, which can lead to reduced lysosomal function. Overall, continuous high glucose reduces microglial and astrocytic ATP production and lysosome activity which may lead to decreased glial oAβ degradation. Our study reveals diabetes-induced hyperglycemia hinders glial oAβ clearance and contributes to oAβ accumulation in AD pathogenesis.
Collapse
Affiliation(s)
- Yung-Cheng Huang
- Department of Physical Medicine and Rehabilitation, Cheng-Hsin General Hospital, Taipei, Taiwan, Republic of China
- National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan, R.O.C
| | - Shu-Meng Hsu
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Feng-Shiun Shie
- Center for Neuropsychiatric Research National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan, R.O.C
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
- Ph.D. Program in Clinical Drug Development of Chinese Herbal Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Li-Jung Chao
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Hui-Wen Chen
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Heng-Hsiang Yao
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Meng An Chien
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Chung-Chih Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- Biophotonics Interdisciplinary Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- * E-mail: (CCL); (HJT)
| | - Huey-Jen Tsay
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- * E-mail: (CCL); (HJT)
| |
Collapse
|
121
|
Dysregulation of mitochondrial dynamics, mitophagy and apoptosis in major depressive disorder: Does inflammation play a role? Mol Psychiatry 2022; 27:1095-1102. [PMID: 34650203 DOI: 10.1038/s41380-021-01312-w] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/07/2021] [Accepted: 09/22/2021] [Indexed: 11/08/2022]
Abstract
Recent studies have suggested that mitochondrial dysfunction and dysregulated neuroinflammatory pathways are involved in the pathophysiology of major depressive disorder (MDD). Here, we aimed to assess the differences in markers of mitochondrial dynamics, mitophagy, general autophagy, and apoptosis in peripheral blood mononuclear cells (PBMCs) of MDD patients (n = 77) and healthy controls (HCs, n = 24). Moreover, we studied inflammation engagement as a moderator of mitochondria dysfunctions on the severity of depressive symptoms. We found increased levels of Mfn-2 (p < 0.001), short Opa-1 (S-Opa-1) (p < 0.001) and Fis-1 (p < 0.001) in MDD patients, suggesting an increase in the mitochondrial fragmentation. We also found that MDD patients had higher levels of Pink-1 (p < 0.001), p62/SQSTM1 (p < 0.001), LC3B (p = 0.002), and caspase-3 active (p = 0.001), and lower levels of parkin (p < 0.001) compared with HCs. Moreover, we showed that that MDD patients with higher CRP levels had higher levels of Mfn-2 (p = 0.001) and LC3B (p = 0.002) when compared with MDD patients with low CRP. Another notable finding was that the severity of depressive symptoms in MDD is associated with changes in protein levels in pathways related to mitochondrial dynamics and mitophagy, and can be dependent on the inflammatory status. Overall, our study demonstrated that a disruption in the mitochondrial dynamics network could initiate a cascade of abnormal changes relevant to the critical pathological changes during the course of MDD and lead to poor outcomes.
Collapse
|
122
|
Gouhar SA, Abo-Elfadl MT, Gamal-Eldeen AM, El-Daly SM. Involvement of miRNAs in response to oxidative stress induced by the steroidal glycoalkaloid α-solanine in hepatocellular carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:212-223. [PMID: 34655286 DOI: 10.1002/tox.23390] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND α-Solanine is a natural toxic glycoalkaloid produced in some species of the Solanaceae family with antiproliferative activity in various cancers. OBJECTIVE This study aimed to investigate the effect of α-solanine on the oxidative stress status in human hepatocellular carcinoma HepG2 cells and to evaluate its influence on microRNAs (miRNAs) associated with oxidative stress and NF-κB regulation. METHODS The prooxidant effect of α-solanine was tested by the decay rate of the fluorescent probe, β-phycoerythrin, and by measuring malondialdehyde, reduced Glutathione, catalase, and superoxide dismutase following treatment of HepG2 cells with low doses of α-solanine. Immunocytochemical techniques were used to detect mitochondrial membrane potential (ΔΨm) and NF-κB protein. The gene expression of NF-κB and miRNAs was evaluated by real-time PCR. RESULTS α-Solanine is a prooxidant that causes a rapid decay in the fluorescence intensity of β-phycoerythrin. It induces oxidative stress-related alterations such as increased lipid peroxidation and reduced antioxidant markers. Oxidative stress induced by α-solanine was mediated by decreased ΔΨm, increased NF-κB expression, upregulation of miRNAs that control oxidative stress by regulating the NF-κB pathway, and downregulation of oncogenic miRNAs that inhibit the NF-κB pathway. CONCLUSION α-Solanine-induced oxidative stress is mediated by alterations in the NF-κB pathway with a detected crosstalk between α-solanine treatment and the expression of oxidative stress-responsive miRNAs.
Collapse
Affiliation(s)
- Shaimaa A Gouhar
- Medical Biochemistry Department, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Mahmoud T Abo-Elfadl
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Cairo, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt
| | - Amira M Gamal-Eldeen
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Cairo, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt
| | - Sherien M El-Daly
- Medical Biochemistry Department, Medical Research Division, National Research Centre, Cairo, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt
| |
Collapse
|
123
|
Zhao M, Lian A, Zhong L, Guo R. The regulatory mechanism between lysosomes and mitochondria in the aetiology of cardiovascular diseases. Acta Physiol (Oxf) 2022; 234:e13757. [PMID: 34978753 DOI: 10.1111/apha.13757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 01/01/2022] [Indexed: 11/28/2022]
Abstract
Coordinated action among various organelles maintains cellular functions. For instance, mitochondria and lysosomes are the main organelles contributing to cellular metabolism and provide energy for cardiomyocyte contraction. They also provide essential signalling platforms in the cell that regulate many key processes such as autophagy, apoptosis, oxidative stress, inflammation and cell death. Often, abnormalities in mitochondrial or lysosomal structures and functions bring about cardiovascular diseases (CVDs). Although the communication between mitochondria and lysosomes throughout the cardiovascular system is intensely studied, the regulatory mechanisms have not been completely understood. Thus, we summarize the most recent studies related to mitochondria and lysosomes' role in CVDs and their potential connections and communications under cardiac pathophysiological conditions. Further, we discuss limitations and future perspectives regarding diagnosis, therapeutic strategies and drug discovery in CVDs.
Collapse
Affiliation(s)
- Mengxue Zhao
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Li Zhong
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Rui Guo
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- The Key Laboratory of Zoological Systematics and Application College of Life Sciences Hebei University Baoding China
| |
Collapse
|
124
|
Kim S, Li L, Zhang J, Jiang C, Lü J. Aqueous metabolome of tissue-specific conditional Pten-knockout mouse prostate cancer and TRAMP neuroendocrine carcinoma. Prostate 2022; 82:154-166. [PMID: 34662447 PMCID: PMC9298286 DOI: 10.1002/pros.24256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Metabolic reprograming is now a recognized hallmark of cancer. The prostate-specific phosphatase and tensin homolog deleted on chromosome 10 (Pten) gene-conditional knockout (KO) mouse carcinogenesis model is highly desirable for studying prostate cancer biology and prevention due to its close resemblance of primary molecular defects and histopathological features of human prostate cancer. We have recently published macromolecular profiling of this model by proteomics and transcriptomics, denoting a preeminence of inflammation and myeloid suppressive immune cell features. Here, we performed metabolomic analyses of Pten-KO prostate versus wild type (WT) counterpart for discernable changes in the aqueous metabolites and contrasted to those in the TRAMP neuroendocrine carcinoma (NECa). METHODS Three matched pairs of tissue-specific conditional Pten-KO mouse prostate and WT prostate of litter/cage-mates at 20-22 weeks of age and three pairs of TRAMP NECa versus WT (28-31 weeks) were profiled for their global aqueous metabolite changes, using hydrophilic interaction liquid chromatography-tandem mass spectrometry. RESULTS The Pten-KO prostate increased purine nucleotide pools, cystathionine, and both reduced and oxidized glutathione (GSH, GSSG), and gluconate/glucuronate species in addition to cholesteryl sulfate and polyamine precursor ornithine. On the contrary, Pten-KO prostate contained diminished pools of glycolytic intermediates and phosphorylcholine derivatives, select amino acids, and their metabolites. Bioinformatic integration revealed a significant shunting of glucose away from glycolysis-citrate cycle and glycerol-lipid genesis to pentose phosphate cycle for NADPH/GSH/GSSG redox and pentose moieties for purine and pyrimidine nucleotides, and glycosylation/glucuronidation. Implicit arginine catabolism to ornithine was consistent with immunosuppression in Pten-KO model. While also increased in cystathionine-GSH/GSSG, purine, and pyrimidine nucleotide pools and glucuronidation at the expense of glycolysis-citrate cycle, the TRAMP NECa increased abundance of many amino acids, methyl donor S-adenosyl-methionine, and intermediates for phospholipids without increasing cholesteryl sulfate or ornithine. CONCLUSIONS The aqueous metabolomic patterns in Pten-KO prostate and TRAMP NECa shared similarities in the greater pools of cystathionine, GSH/GSSG redox pair, and nucleotides and shunting away from glycolysis-citrate cycle in both models. Remarkable metabolic distinctions between them included metabolisms of many amino acids (protein synthesis; arginine-ornithine/immune suppression) and cholesteryl sulfate and methylation donor for epigenetic regulations.
Collapse
Affiliation(s)
- Sangyub Kim
- Department of PharmacologyPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Present address:
Sangyub Kim, Zentalis PharmaceuticalsSan DiegoCaliforniaUSA
| | - Li Li
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
- Present address:
Li Li and Jinhui Zhang, Food and Drug AdministrationSilver SpringMarylandUSA
| | - Jinhui Zhang
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
- Present address:
Li Li and Jinhui Zhang, Food and Drug AdministrationSilver SpringMarylandUSA
| | - Cheng Jiang
- Department of PharmacologyPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
| | - Junxuan Lü
- Department of PharmacologyPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
| |
Collapse
|
125
|
Sarkar A, Hameed R, Mishra A, Bhatta RS, Nazir A. Genetic modulators associated with regulatory surveillance of mitochondrial quality control, play a key role in regulating stress pathways and longevity in C. elegans. Life Sci 2021; 290:120226. [PMID: 34953889 DOI: 10.1016/j.lfs.2021.120226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/22/2021] [Accepted: 12/08/2021] [Indexed: 12/21/2022]
Abstract
The multi-factorial Parkinson's disease (PD) is known to be associated with mitochondrial dysfunction, endoplasmic reticulum stress, alpha synuclein aggregation and dopaminergic neuronal death, with oxidative stress being a common denominator to these underlying processes. The perception of mitochondria being 'just ATP producing compartments' have been counterpoised as studies, particularly related to PD, have underlined their strong role in cause and progression of the disease. During PD pathogenesis, neurons encounter chronic stress conditions mainly due to failure of Mitochondrial Quality Control (MQC) machinery. To dissect the regulatory understanding of mitochondrial dysfunction during neurological disease progression, we endeavored to identify key regulatory endpoints that control multiple facets of MQC machinery. Our studies, employing transgenic C. elegans strain expressing human α-synuclein, led us to identification of mitochondrial genes nuo-5 (involved in oxidative phosphorylation), F25B4.7 (exhibits ATP transmembrane transporter activity) and C05D11.9 (having ribonuclease activity), which form predicted downstream targets of most elevated and down-regulated mi-RNA molecules. RNAi mediated silencing, gene ontology and functional genomics analysis studies demonstrated their role in modulating major MQC pathways. The attenuated MQC pathways mainly affected clearance of misfolded and aggregated proteins, redox homeostasis and longevity with compromised dopaminergic functions. Overexpression of the mitochondrial genes by 3 beta-hydroxyl steroid, Tomatidine, was found to curtail the redox imbalance thus leading to amelioration of effects associated with PD and an increase in the lifespan of treated nematodes. Therefore, this study unveils the regulatory role of mitochondrial genes as critical modulators of stress control involved in effects associated with PD pathogenesis.
Collapse
Affiliation(s)
- Arunabh Sarkar
- Division of Neuroscience and Aging Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Rohil Hameed
- Division of Neuroscience and Aging Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Anjali Mishra
- Division of Neuroscience and Aging Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Rabi Sankar Bhatta
- Division of Neuroscience and Aging Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Aamir Nazir
- Division of Neuroscience and Aging Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP, India.
| |
Collapse
|
126
|
Bonam SR, Tranchant C, Muller S. Autophagy-Lysosomal Pathway as Potential Therapeutic Target in Parkinson's Disease. Cells 2021; 10:3547. [PMID: 34944054 PMCID: PMC8700067 DOI: 10.3390/cells10123547] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
Cellular quality control systems have gained much attention in recent decades. Among these, autophagy is a natural self-preservation mechanism that continuously eliminates toxic cellular components and acts as an anti-ageing process. It is vital for cell survival and to preserve homeostasis. Several cell-type-dependent canonical or non-canonical autophagy pathways have been reported showing varying degrees of selectivity with regard to the substrates targeted. Here, we provide an updated review of the autophagy machinery and discuss the role of various forms of autophagy in neurodegenerative diseases, with a particular focus on Parkinson's disease. We describe recent findings that have led to the proposal of therapeutic strategies targeting autophagy to alter the course of Parkinson's disease progression.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Christine Tranchant
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France;
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, 67400 Illkirch, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Sylviane Muller
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
- CNRS and Strasbourg University, Unit Biotechnology and Cell Signaling/Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), 67000 Strasbourg, France
| |
Collapse
|
127
|
Yapici N, Gao X, Yan X, Hou S, Jockusch S, Lesniak L, Gibson KM, Bi L. Novel Dual-Organelle-Targeting Probe (RCPP) for Simultaneous Measurement of Organellar Acidity and Alkalinity in Living Cells. ACS OMEGA 2021; 6:31447-31456. [PMID: 34869971 PMCID: PMC8637586 DOI: 10.1021/acsomega.1c03087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/01/2021] [Indexed: 05/06/2023]
Abstract
Many organelles, such as lysosomes and mitochondria, maintain a pH that is different from the cytoplasmic pH. These pH differences have important functional ramifications for those organelles. Many cellular events depend upon a well-compartmentalized distribution of H+ ions spanning the membrane for the optimal function. Cells have developed a variety of mechanisms that enable the regulation of organelle pH. However, the measurement of organellar acidity/alkalinity in living cells has remained a challenge. Currently, most existing probes for the estimation of intracellular pH show a single -organelle targeting capacity. Such probes provide data that fails to comprehensively reveal the pathological and physiological roles and connections between mitochondria and lysosomes in different species. Mitochondrial and lysosomal functions are closely related and important for regulating cellular homeostasis. Accordingly, the design of a single fluorescent probe that can simultaneously target mitochondria and lysosomes is highly desirable, enabling a better understanding of the crosstalk between these organelles. We report the development of a novel fluorescent sensor, rhodamine-coumarin pH probe (RCPP), for detection of organellar acidity/alkalinity. RCPP simultaneously moves between mitochondrion and lysosome subcellular locations, facilitating the simultaneous monitoring of pH alterations in mitochondria and lysosomes.
Collapse
Affiliation(s)
- Nazmiye
B. Yapici
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Xiang Gao
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Xin Yan
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Shanshan Hou
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Steffen Jockusch
- Department
of Chemistry, Columbia University, New York, New York 10027, United States
| | - Lillian Lesniak
- Department
of Chemical Engineering, Michigan Technological
University, Houghton, Michigan 49931, United States
| | - K. Michael Gibson
- Department
of Pharmacotherapy, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Lanrong Bi
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
128
|
Hello from the other side: Membrane contact of lipid droplets with other organelles and subsequent functional implications. Prog Lipid Res 2021; 85:101141. [PMID: 34793861 DOI: 10.1016/j.plipres.2021.101141] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Lipid droplets (LDs) are ubiquitous organelles that play crucial roles in response to physiological and environmental cues. The identification of several neutral lipid synthesizing and regulatory protein complexes have propelled significant advance on the mechanisms of LD biogenesis in the endoplasmic reticulum (ER). Increasing evidence suggests that distinct proteins and regulatory factors, which localize to membrane contact sites (MCS), are involved not only in interorganellar lipid exchange and transport, but also function in other important cellular processes, including autophagy, mitochondrial dynamics and inheritance, ion signaling and inter-regulation of these MCS. More and more tethers and molecular determinants are associated to MCS and to a diversity of cellular and pathophysiological processes, demonstrating the dynamics and importance of these junctions in health and disease. The conjugation of lipids with proteins in supramolecular complexes is known to be paramount for many biological processes, namely membrane biosynthesis, cell homeostasis, regulation of organelle division and biogenesis, and cell growth. Ultimately, this physical organization allows the contact sites to function as crucial metabolic hubs that control the occurrence of chemical reactions. This leads to biochemical and metabolite compartmentalization for the purposes of energetic efficiency and cellular homeostasis. In this review, we will focus on the structural and functional aspects of LD-organelle interactions and how they ensure signaling exchange and metabolites transfer between organelles.
Collapse
|
129
|
Bertman KA, Abeywickrama CS, Pang Y. A NIR Emitting Cyanine with Large Stokes' Shift for Mitochondria and Identification of their Membrane Potential Disruption. Chembiochem 2021; 23:e202100516. [PMID: 34783144 DOI: 10.1002/cbic.202100516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/12/2021] [Indexed: 12/11/2022]
Abstract
An NIR emitting (λem ≈730 nm) cyanine probe ExCy was synthesized in good yields by extending the π-conjugation length (i. e., with furan moiety) to the donor-accepter system. ExCy exhibited a large Stokes' shift (Δλ≈100 nm) due to strong intramolecular charge transfer (ICT), and high fluorescence quantum yield (Φfl ≈0.47 in DCM). Due to its low fluorescence in an aqueous environment (Φfl ≈0.007 in H2 O), the probe exhibited the potential of achieving a large fluorescence turn-on upon entering a hydrophobic cellular environment. Fluorescence confocal microscopy studies revealed that ExCy was readily excitable with a far-red laser line (i. e., 640 nm) while the corresponding emission was collected in the NIR region. ExCy exhibited excellent selectivity towards live cell mitochondria according to the co-localization studies. The probe also exhibited high photostability, long-term imaging ability and wash-free staining ability, when being applied to live cells. Our studies indicated that the mitochondrial localization of ExCy was dependent on the membrane potential of the mitochondria. ExCy was successfully utilized as a mitochondrial membrane potential dysfunction indicator to visually identify cells with mitochondrial dysfunction via fluorescence confocal microscopy. ExCy was further examined for potential in vivo imaging of zebrafish.
Collapse
Affiliation(s)
- Keti A Bertman
- Department of Chemistry, University of Akron, Akron, Ohio, 44325, USA
| | | | - Yi Pang
- Department of Chemistry, University of Akron, Akron, Ohio, 44325, USA.,Maurice Morton Institute of Polymer Science, University of Akron, Akron, Ohio, 44325, USA
| |
Collapse
|
130
|
Constitutive activation of the PI3K-Akt-mTORC1 pathway sustains the m.3243 A > G mtDNA mutation. Nat Commun 2021; 12:6409. [PMID: 34737295 PMCID: PMC8568893 DOI: 10.1038/s41467-021-26746-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/20/2021] [Indexed: 11/25/2022] Open
Abstract
Mutations of the mitochondrial genome (mtDNA) cause a range of profoundly debilitating clinical conditions for which treatment options are very limited. Most mtDNA diseases show heteroplasmy – tissues express both wild-type and mutant mtDNA. While the level of heteroplasmy broadly correlates with disease severity, the relationships between specific mtDNA mutations, heteroplasmy, disease phenotype and severity are poorly understood. We have carried out extensive bioenergetic, metabolomic and RNAseq studies on heteroplasmic patient-derived cells carrying the most prevalent disease related mtDNA mutation, the m.3243 A > G. These studies reveal that the mutation promotes changes in metabolites which are associated with the upregulation of the PI3K-Akt-mTORC1 axis in patient-derived cells and tissues. Remarkably, pharmacological inhibition of PI3K, Akt, or mTORC1 reduced mtDNA mutant load and partially rescued cellular bioenergetic function. The PI3K-Akt-mTORC1 axis thus represents a potential therapeutic target that may benefit people suffering from the consequences of the m.3243 A > G mutation. Heteroplasmic mtDNA mutations cause disease in humans. Here, Chung et al find the PI3K-Akt-mTORC1 pathway constitutively activated in cells with the heteroplasmic m.3243 A > G mutation, and inhibition of the pathway cell autonomously reduces mutant mtDNA load and rescues mitochondrial bioenergetics.
Collapse
|
131
|
Beyer AM, Norwood Toro LE, Hughes WE, Young M, Clough AV, Gao F, Medhora M, Audi SH, Jacobs ER. Autophagy, TERT, and mitochondrial dysfunction in hyperoxia. Am J Physiol Heart Circ Physiol 2021; 321:H985-H1003. [PMID: 34559580 PMCID: PMC8616608 DOI: 10.1152/ajpheart.00166.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/31/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023]
Abstract
Ventilation with gases containing enhanced fractions of oxygen is the cornerstone of therapy for patients with hypoxia and acute respiratory distress syndrome. Yet, hyperoxia treatment increases free reactive oxygen species (ROS)-induced lung injury, which is reported to disrupt autophagy/mitophagy. Altered extranuclear activity of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), plays a protective role in ROS injury and autophagy in the systemic and coronary endothelium. We investigated interactions between autophagy/mitophagy and TERT that contribute to mitochondrial dysfunction and pulmonary injury in cultured rat lung microvascular endothelial cells (RLMVECs) exposed in vitro, and rat lungs exposed in vivo to hyperoxia for 48 h. Hyperoxia-induced mitochondrial damage in rat lungs [TOMM20, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], which was paralleled by increased markers of inflammation [myeloperoxidase (MPO), IL-1β, TLR9], impaired autophagy signaling (Beclin-1, LC3B-II/1, and p62), and decreased the expression of TERT. Mitochondrial-specific autophagy (mitophagy) was not altered, as hyperoxia increased expression of Pink1 but not Parkin. Hyperoxia-induced mitochondrial damage (TOMM20) was more pronounced in rats that lack the catalytic subunit of TERT and resulted in a reduction in cellular proliferation rather than cell death in RLMVECs. Activation of TERT or autophagy individually offset mitochondrial damage (MTT). Combined activation/inhibition failed to alleviate hyperoxic-induced mitochondrial damage in vitro, whereas activation of autophagy in vivo decreased mitochondrial damage (MTT) in both wild type (WT) and rats lacking TERT. Functionally, activation of either TERT or autophagy preserved transendothelial membrane resistance. Altogether, these observations show that activation of autophagy/mitophagy and/or TERT mitigate loss of mitochondrial function and barrier integrity in hyperoxia.NEW & NOTEWORTHY In cultured pulmonary artery endothelial cells and in lungs exposed in vivo to hyperoxia, autophagy is activated, but clearance of autophagosomes is impaired in a manner that suggests cross talk between TERT and autophagy. Stimulation of autophagy prevents hyperoxia-induced decreases in mitochondrial metabolism and sustains monolayer resistance. Hyperoxia increases mitochondrial outer membrane (TOMM20) protein, decreases mitochondrial function, and reduces cellular proliferation without increasing cell death.
Collapse
Affiliation(s)
- Andreas M Beyer
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Laura E Norwood Toro
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - William E Hughes
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Micaela Young
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anne V Clough
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
- Department of Mathematics, Statistics and Computer Science, Marquette University, Milwaukee, Wisconsin
| | - Feng Gao
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Meetha Medhora
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
| | - Said H Audi
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
- Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin
| | - Elizabeth R Jacobs
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
| |
Collapse
|
132
|
Huang YL, Pan WL, Cai WW, Ju JQ, Sun SC. Exposure to citrinin induces DNA damage, autophagy, and mitochondria dysfunction during first cleavage of mouse embryos. ENVIRONMENTAL TOXICOLOGY 2021; 36:2217-2224. [PMID: 34314561 DOI: 10.1002/tox.23335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/13/2021] [Accepted: 07/17/2021] [Indexed: 06/13/2023]
Abstract
Citrinin (CTN) is a mycotoxin, which is isolated from Penicillium citrinum and widely existed in the contaminated feeds. It is reported that CTN is toxic to heart, liver, and reproductive system. Previous studies indicated that CTN induced apoptosis in oocytes and embryos. In this study, we reported the potential causes of CTN on embryo development. Our results showed that 40 μM CTN exposure significantly reduced the first cleavage of mouse embryos, showing with the low rate of 2-cell embryos. We found that CTN induced DNA damage, showing the higher positive γH2A.X signals. Autophagy was occurred since more LC3 positive autophagosomes were found in the cytoplasm. This could be confirmed by the enhanced lysosome function, since higher accumulated lysosome distribution were found and LAMP2 was also increased under CTN exposure. Besides, we showed that mitochondria distribution was disturbed, indicating that CTN could disrupt mitochondria function, which could be the possible reason for the oxidative stress and apoptosis in CTN-exposed embryos. In conclusion, our study showed that CTN exposure had adverse effects on the early embryo development during first cleavage through its effects on the induction of DNA damage, autophagy, and mitochondria dysfunction.
Collapse
Affiliation(s)
- Yi-Lin Huang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wen-Lin Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wen-Wu Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
133
|
Gutiérrez-Pérez P, Santillán EM, Lendl T, Wang J, Schrempf A, Steinacker TL, Asparuhova M, Brandstetter M, Haselbach D, Cochella L. miR-1 sustains muscle physiology by controlling V-ATPase complex assembly. SCIENCE ADVANCES 2021; 7:eabh1434. [PMID: 34652942 PMCID: PMC8519577 DOI: 10.1126/sciadv.abh1434] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/23/2021] [Indexed: 05/13/2023]
Abstract
Muscle function requires unique structural and metabolic adaptations that can render muscle cells selectively vulnerable, with mutations in some ubiquitously expressed genes causing myopathies but sparing other tissues. We uncovered a muscle cell vulnerability by studying miR-1, a deeply conserved, muscle-specific microRNA whose ablation causes various muscle defects. Using Caenorhabditis elegans, we found that miR-1 represses multiple subunits of the ubiquitous vacuolar adenosine triphosphatase (V-ATPase) complex, which is essential for internal compartment acidification and metabolic signaling. V-ATPase subunits are predicted miR-1 targets in animals ranging from C. elegans to humans, and we experimentally validated this in Drosophila. Unexpectedly, up-regulation of V-ATPase subunits upon miR-1 deletion causes reduced V-ATPase function due to defects in complex assembly. These results reveal V-ATPase assembly as a conserved muscle cell vulnerability and support a previously unknown role for microRNAs in the regulation of protein complexes.
Collapse
Affiliation(s)
- Paula Gutiérrez-Pérez
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Emilio M. Santillán
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Thomas Lendl
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Jingkui Wang
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Anna Schrempf
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | | | - Mila Asparuhova
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Marlene Brandstetter
- Electron Microscopy Facility, Vienna BioCenter Core Facilities GmbH, Vienna, Austria
| | - David Haselbach
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Luisa Cochella
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| |
Collapse
|
134
|
Shyam R, Ogando DG, Choi M, Liton PB, Bonanno JA. Mitochondrial ROS Induced Lysosomal Dysfunction and Autophagy Impairment in an Animal Model of Congenital Hereditary Endothelial Dystrophy. Invest Ophthalmol Vis Sci 2021; 62:15. [PMID: 34533563 PMCID: PMC8458782 DOI: 10.1167/iovs.62.12.15] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/18/2021] [Indexed: 01/04/2023] Open
Abstract
Purpose The Slc4a11 knock out (KO) mouse model recapitulates the human disease phenotype associated with congenital hereditary endothelial dystrophy (CHED). Increased mitochondrial reactive oxygen species (ROS) in the Slc4a11 KO mouse model is a major cause of edema and endothelial cell loss. Here, we asked if autophagy was activated by ROS in the KO mice. Methods Immortalized cell lines and mouse corneal endothelia were used to measure autophagy and lysosome associated protein expressions using Protein Simple Wes immunoassay. Autophagy and lysosome functions were examined in wild type (WT) and KO cells as well as animals treated with the mitochondrial ROS quencher MitoQ. Results Even though autophagy activation was evident, autophagy flux was aberrant in Slc4a11 KO cells and corneal endothelium. Expression of lysosomal proteins and lysosomal mass were decreased along with reduced nuclear translocation of lysosomal master regulator, transcription factor EB (TFEB). MitoQ reversed aberrant lysosomal functions and TFEB nuclear localization in KO cells. MitoQ injections in KO animals reduced corneal edema and decreased the rate of endothelial cell loss. Conclusions Mitochondrial ROS disrupts TFEB signaling causing lysosomal dysfunction with impairment of autophagy in Slc4a11 KO corneal endothelium. Our study is the first to identify the presence as well as cause of lysosomal dysfunction in an animal model of CHED, and to identify a potential therapeutic approach.
Collapse
MESH Headings
- Animals
- Anion Transport Proteins/genetics
- Autophagy/physiology
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Blotting, Western
- Cathepsin L/metabolism
- Cells, Cultured
- Corneal Dystrophies, Hereditary/genetics
- Corneal Dystrophies, Hereditary/metabolism
- Corneal Dystrophies, Hereditary/pathology
- Disease Models, Animal
- Endothelium, Corneal/drug effects
- Endothelium, Corneal/metabolism
- Gene Expression Regulation
- Immunohistochemistry
- Injections, Intraperitoneal
- Lysosomes/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Fluorescence
- Mitochondria/metabolism
- Organophosphorus Compounds/pharmacology
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Symporters/genetics
- Transfection
- Ubiquinone/analogs & derivatives
- Ubiquinone/pharmacology
Collapse
Affiliation(s)
- Rajalekshmy Shyam
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Diego G. Ogando
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Moonjung Choi
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Paloma B. Liton
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Joseph A. Bonanno
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| |
Collapse
|
135
|
Abeywickrama CS, Baumann HJ, Pang Y. Simultaneous Visualization of Mitochondria and Lysosome by a Single Cyanine Dye: The Impact of the Donor Group (-NR 2) Towards Organelle Selectivity. J Fluoresc 2021; 31:1227-1234. [PMID: 34297321 DOI: 10.1007/s10895-021-02786-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/13/2021] [Indexed: 11/29/2022]
Abstract
A benzothiazolium-based hemicyanine dye (probe 3) has been synthesized by attaching a morpholine group into a phenyl benzothiazolium skeleton. Probe 3 exhibited interesting photophysical characteristics including red emission (λem ≈600 nm), enhanced Stokes shift (Δλ ≈80 nm) and sensitivity to solvent polarity. Although the probe 3 exhibited almost no emission in aqueous environments (φfl ≈0.002), its fluorescence could be increased by ≈50 fold in organic solvents (φfl ≈0.10), making it possible for live cell imaging under wash-free conditions. Probe 3 exhibited excellent ability to visualize cellular mitochondria and lysosomes simultaneously, as observed from fluorescence confocal microscopy. In addition, probe 3 also exhibited good biocompatibility (calculated LC50 > 20 µM) and high photostability.
Collapse
Affiliation(s)
- Chathura S Abeywickrama
- Department of Chemistry and Maurice Morton Institute of Polymer Science, University of Akron, Akron, OH, 44325, USA
| | - Hannah J Baumann
- Department of Chemistry and Maurice Morton Institute of Polymer Science, University of Akron, Akron, OH, 44325, USA
| | - Yi Pang
- Department of Chemistry and Maurice Morton Institute of Polymer Science, University of Akron, Akron, OH, 44325, USA.
| |
Collapse
|
136
|
Zhou S, Ling X, Meng P, Liang Y, Shen K, Wu Q, Zhang Y, Chen Q, Chen S, Liu Y, Zhou L. Cannabinoid receptor 2 plays a central role in renal tubular mitochondrial dysfunction and kidney ageing. J Cell Mol Med 2021; 25:8957-8972. [PMID: 34414658 PMCID: PMC8435409 DOI: 10.1111/jcmm.16857] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/24/2021] [Accepted: 07/31/2021] [Indexed: 12/13/2022] Open
Abstract
Kidney is one of the most important organs in maintaining the normal life activities. With the high abundance of mitochondria, renal tubular cell plays the vital role in functioning in the reabsorption and secretion of kidney. Reports have shown that mitochondrial dysfunction is of great importance to renal tubular cell senescence and subsequent kidney ageing. However, the underlying mechanisms are not elucidated. Cannabinoid receptor 2 is one of the two receptors responsible for the activation of endocannabinoid system. CB2 is primarily upregulated in renal tubular cells in chronic kidney diseases and mediates fibrogenesis. However, the role of CB2 in tubular mitochondrial dysfunction and kidney ageing has not been clarified. In this study, we found that CB2 was upregulated in kidneys in 24-month-old mice and d-galactose (d-gal)-induced accelerated ageing mice, accompanied by the decrease in mitochondrial mass. Furthermore, gene deletion of CB2 in d-gal-treated mice could greatly inhibit the activation of β-catenin signalling and restore the mitochondrial integrity and Adenosine triphosphate (ATP) production. In CB2 knockout mice, renal tubular cell senescence and kidney fibrosis were also significantly inhibited. CB2 overexpression or activation by the agonist AM1241 could sufficiently induce the decrease in PGC-1α and a variety of mitochondria-related proteins and trigger cellular senescence in cultured human renal proximal tubular cells. CB2-activated mitochondrial dysfunction and cellular senescence could be blocked by ICG-001, a blocker for β-catenin signalling. These results show CB2 plays a central role in renal tubular mitochondrial dysfunction and kidney ageing. The intrinsic mechanism may be related to its activation in β-catenin signalling.
Collapse
Affiliation(s)
- Shan Zhou
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xian Ling
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ping Meng
- Department of Central LaboratoryHuadu District People’s HospitalSouthern Medical UniversityGuangzhouChina
| | - Ye Liang
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Kunyu Shen
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Qinyu Wu
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yunfang Zhang
- Department of Nephrologythe First People's Hospital of FoshanFoshanChina
| | - Qiyan Chen
- Department of Nephrologythe First People's Hospital of FoshanFoshanChina
| | - Shuangqin Chen
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Youhua Liu
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Lili Zhou
- State Key Laboratory of Organ Failure ResearchNational Clinical Research Center of Kidney DiseaseDivision of NephrologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
137
|
Deus CM, Pereira SP, Cunha-Oliveira T, Teixeira J, Simões RF, Cagide F, Benfeito S, Borges F, Raimundo N, Oliveira PJ. A mitochondria-targeted caffeic acid derivative reverts cellular and mitochondrial defects in human skin fibroblasts from male sporadic Parkinson's disease patients. Redox Biol 2021; 45:102037. [PMID: 34147843 PMCID: PMC8220403 DOI: 10.1016/j.redox.2021.102037] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 12/24/2022] Open
Abstract
Parkinson's Disease (PD) is a neurodegenerative disorder affecting more than 10 million people worldwide. Currently, PD has no cure and no early diagnostics methods exist. Mitochondrial dysfunction is presented in the early stages of PD, and it is considered an important pathophysiology component. We have previously developed mitochondria-targeted hydroxycinnamic acid derivatives, presenting antioxidant and iron-chelating properties, and preventing oxidative stress in several biological models of disease. We have also demonstrated that skin fibroblasts from male sporadic PD patients (sPD) presented cellular and mitochondrial alterations, including increased oxidative stress, hyperpolarized and elongated mitochondria and decreased respiration and ATP levels. We also showed that forcing mitochondrial oxidative phosphorylation (OXPHOS) in sPD fibroblasts uncovers metabolic defects that were otherwise hidden. In this work, we tested the hypothesis that a lead mitochondria-targeted hydroxycinnamic acid derivative would revert the phenotype found in skin fibroblasts from sPD patients. Our results demonstrated that treating human skin fibroblasts from sPD patients with non-toxic concentrations of AntiOxCIN4 restored mitochondrial membrane potential and mitochondrial fission, decreased autophagic flux, and enhanced cellular responses to stress by improving the cellular redox state and decreasing reactive oxygen species (ROS) levels. Besides, fibroblasts from sPD patients treated with AntiOxCIN4 showed increased maximal respiration and metabolic activity, converting sPD fibroblasts physiologically more similar to their sex- and age-matched healthy controls. The positive compound effect was reinforced using a supervised machine learning model, confirming that AntiOxCIN4 treatment converted treated fibroblasts from sPD patients closer to the phenotype of control fibroblasts. Our data points out a possible mechanism of AntiOxCIN4 action contributing to a deeper understanding of how the use of mitochondria-targeted antioxidants based on a polyphenol scaffold can be used as potential drug candidates for delaying PD progression, validating the use of fibroblasts from sPD patients with more active OXPHOS as platforms for mitochondria-based drug development.
Collapse
Affiliation(s)
- Cláudia M Deus
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Susana P Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Research Centre in Physical Activity Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, Porto, Portugal
| | - Teresa Cunha-Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - José Teixeira
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rui F Simões
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Fernando Cagide
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Sofia Benfeito
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Nuno Raimundo
- Penn State University College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA; Multidisciplinary Institute of Ageing (MIA), University of Coimbra, Coimbra, Portugal
| | - Paulo J Oliveira
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
138
|
Zuo SQ, Li C, Liu YL, Tan YH, Wan X, Xu T, Li Q, Wang L, Wu YL, Deng FM, Tang B. Cordycepin inhibits cell senescence by ameliorating lysosomal dysfunction and inducing autophagy through the AMPK and mTOR-p70S6K pathway. FEBS Open Bio 2021; 11:2705-2714. [PMID: 34448542 PMCID: PMC8487049 DOI: 10.1002/2211-5463.13263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/10/2021] [Accepted: 08/02/2021] [Indexed: 01/10/2023] Open
Abstract
Cell senescence is closely related to autophagy. In this article, we identified a natural nucleoside analogue, cordycepin, that has the ability to significantly improve lysosomal function, enhance the activity of the lysosomal representative protease cathepsin B (CTSB), and promote the expression of the functional protein lysosomal‐associated membrane protein 2 (LAMP2) on the lysosomal membrane. Cordycepin then restores the damaged autophagy level of aging cells by activating the classic AMPK and mTOR–p70S6K signaling pathways, thus inhibiting cell senescence in an H2O2‐induced stress‐induced premature senescence (SIPS) cell model. This study provides new theoretical support for the further development of cordycepin and clinical antiaging drugs to inhibit cell senescence and suggests that the regulatory mechanisms of lysosomes in senescent cells should be considered when treating age‐related diseases.
Collapse
Affiliation(s)
- Shi Qi Zuo
- School of Clinical Medicine, Chengdu Medical College, China
| | - Can Li
- School of Basic Medical Science, Chengdu Medical College, China.,Sichuan Clinical Research Center for Geriatrics, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, China
| | - Yi Lun Liu
- Sichuan Clinical Research Center for Geriatrics, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, China.,People's Hospital of Mingshan District, Ya'an, China
| | - Yue Hao Tan
- School of Basic Medical Science, Chengdu Medical College, China
| | - Xing Wan
- School of Clinical Medicine, Chengdu Medical College, China
| | - Tian Xu
- Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Qiang Li
- Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Li Wang
- Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li Wu
- Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Feng Mei Deng
- School of Basic Medical Science, Chengdu Medical College, China
| | - Bin Tang
- School of Basic Medical Science, Chengdu Medical College, China
| |
Collapse
|
139
|
Burtscher J, Syed MMK, Keller MA, Lashuel HA, Millet GP. Fatal attraction - The role of hypoxia when alpha-synuclein gets intimate with mitochondria. Neurobiol Aging 2021; 107:128-141. [PMID: 34428721 DOI: 10.1016/j.neurobiolaging.2021.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022]
Abstract
Alpha-synuclein aggregation and mitochondrial dysfunction are main pathological hallmarks of Parkinson's disease (PD) and several other neurodegenerative diseases, collectively known as synucleinopathies. However, increasing evidence suggests that they may not be sufficient to cause PD. Here we propose the role of hypoxia as a missing link that connects the complex interplay between alpha-synuclein biochemistry and pathology, mitochondrial dysfunctions and neurodegeneration in PD. We review the partly conflicting literature on alpha-synuclein binding to membranes and mitochondria and its impact on mitochondrial functions. From there, we focus on adverse changes in cellular environments, revolving around hypoxic stress, that may trigger or facilitate PD progression. Inter-dependent structural re-arrangements of mitochondrial membranes, including increased cytoplasmic exposure of mitochondrial cardiolipins and changes in alpha-synuclein localization and conformation are discussed consequences of such conditions. Enhancing cellular resilience could be an integral part of future combination-based therapies of PD. This may be achieved by boosting the capacity of cellular and specifically mitochondrial processes to regulate and adapt to altered proteostasis, redox, and inflammatory conditions and by inducing protective molecular and tissue re-modelling.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland; Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland.
| | - Muhammed Muazzam Kamil Syed
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
140
|
Sanchez-Alvarez NT, Bautista-Niño PK, Trejos-Suárez J, Serrano-Diaz NC. Metachromatic Leukodystrophy: Diagnosis and Treatment Challenges. BIONATURA 2021. [DOI: 10.21931/rb/2021.06.03.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Metachromatic leukodystrophy is a neurological disease of the lysosomal deposit that has a significant impact given the implications for the neurodegenerative deterioration of the patient. Currently, there is no treatment available that reverses the development of characteristic neurological and systemic symptoms. Objective. Carry out an updated bibliographic search on the most critical advances in the treatment and diagnosis for LDM. A retrospective topic review published in English and Spanish in the Orphanet and Pubmed databases. Current treatment options, such as enzyme replacement therapy and hematopoietic stem cell transplantation aimed at decreasing the rapid progression of the disease, improving patient survival; however, these are costly. The pathophysiological events of intracellular signaling related to the deficiency of the enzyme Arylsulfatase A and subsequent accumulation of sulphatides and glycosylated ceramides have not yet been established. Recently, the accumulation of C16 sulphatides has been shown to inhibit glycolysis and insulin secretion in pancreatic cells. The significant advance in technology has allowed timely diagnosis in patients suffering from LDM; however, they still do not have an effective treatment.
Collapse
Affiliation(s)
- Nayibe Tatiana Sanchez-Alvarez
- Universidad del Valle, Faculty of Health, Biomedical Sciences Doctorate Program, Colombian Cardiovascular Foundation, Research Center. Floridablanca, Santander, Colombia. Universidad de Santander, Faculty of Health Sciences, CliniUDES Research Group, Bucaramanga, Santander, Colombia
| | | | - Juanita Trejos-Suárez
- Universidad de Santander, Faculty of Health Sciences, CliniUDES Research Group, Bucaramanga, Santander, Colombia
| | | |
Collapse
|
141
|
Rizzollo F, More S, Vangheluwe P, Agostinis P. The lysosome as a master regulator of iron metabolism. Trends Biochem Sci 2021; 46:960-975. [PMID: 34384657 DOI: 10.1016/j.tibs.2021.07.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022]
Abstract
Intracellular iron fulfills crucial cellular processes, including DNA synthesis and mitochondrial metabolism, but also mediates ferroptosis, a regulated form of cell death driven by lipid-based reactive oxygen species (ROS). Beyond their established role in degradation and recycling, lysosomes occupy a central position in iron homeostasis and integrate metabolic and cell death signals emanating from different subcellular sites. We discuss the central role of the lysosome in preserving iron homeostasis and provide an integrated outlook of the regulatory circuits coupling the lysosomal system to the control of iron trafficking, interorganellar crosstalk, and ferroptosis induction. We also discuss novel studies unraveling how deregulated lysosomal iron-handling functions contribute to cancer, neurodegeneration, and viral infection, and can be harnessed for therapeutic interventions.
Collapse
Affiliation(s)
- Francesca Rizzollo
- Laboratory of Cell Death and Research, Vlaams Instituut voor Biotechnologie (VIB)-Katholieke Universiteit (KU) Leuven Center for Cancer Biology, Leuven, Belgium; Laboratory of Cell Death and Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sanket More
- Laboratory of Cell Death and Research, Vlaams Instituut voor Biotechnologie (VIB)-Katholieke Universiteit (KU) Leuven Center for Cancer Biology, Leuven, Belgium; Laboratory of Cell Death and Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| | - Patrizia Agostinis
- Laboratory of Cell Death and Research, Vlaams Instituut voor Biotechnologie (VIB)-Katholieke Universiteit (KU) Leuven Center for Cancer Biology, Leuven, Belgium; Laboratory of Cell Death and Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
142
|
Liu M, Wu N, Xu K, Saaoud F, Vasilopoulos E, Shao Y, Zhang R, Wang J, Shen H, Yang WY, Lu Y, Sun Y, Drummer C, Liu L, Li L, Hu W, Yu J, Praticò D, Sun J, Jiang X, Wang H, Yang X. Organelle Crosstalk Regulators Are Regulated in Diseases, Tumors, and Regulatory T Cells: Novel Classification of Organelle Crosstalk Regulators. Front Cardiovasc Med 2021; 8:713170. [PMID: 34368262 PMCID: PMC8339352 DOI: 10.3389/fcvm.2021.713170] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022] Open
Abstract
To examine whether the expressions of 260 organelle crosstalk regulators (OCRGs) in 16 functional groups are modulated in 23 diseases and 28 tumors, we performed extensive -omics data mining analyses and made a set of significant findings: (1) the ratios of upregulated vs. downregulated OCRGs are 1:2.8 in acute inflammations, 1:1 in metabolic diseases, 1:1.2 in autoimmune diseases, and 1:3.8 in organ failures; (2) sepsis and trauma-upregulated OCRG groups such as vesicle, mitochondrial (MT) fission, and mitophagy but not others, are termed as the cell crisis-handling OCRGs. Similarly, sepsis and trauma plus organ failures upregulated seven OCRG groups including vesicle, MT fission, mitophagy, sarcoplasmic reticulum–MT, MT fusion, autophagosome–lysosome fusion, and autophagosome/endosome–lysosome fusion, classified as the cell failure-handling OCRGs; (3) suppression of autophagosome–lysosome fusion in endothelial and epithelial cells is required for viral replications, which classify this decreased group as the viral replication-suppressed OCRGs; (4) pro-atherogenic damage-associated molecular patterns (DAMPs) such as oxidized low-density lipoprotein (oxLDL), lipopolysaccharide (LPS), oxidized-1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (oxPAPC), and interferons (IFNs) totally upregulated 33 OCRGs in endothelial cells (ECs) including vesicle, MT fission, mitophagy, MT fusion, endoplasmic reticulum (ER)–MT contact, ER– plasma membrane (PM) junction, autophagosome/endosome–lysosome fusion, sarcoplasmic reticulum–MT, autophagosome–endosome/lysosome fusion, and ER–Golgi complex (GC) interaction as the 10 EC-activation/inflammation-promoting OCRG groups; (5) the expression of OCRGs is upregulated more than downregulated in regulatory T cells (Tregs) from the lymph nodes, spleen, peripheral blood, intestine, and brown adipose tissue in comparison with that of CD4+CD25− T effector controls; (6) toll-like receptors (TLRs), reactive oxygen species (ROS) regulator nuclear factor erythroid 2-related factor 2 (Nrf2), and inflammasome-activated regulator caspase-1 regulated the expressions of OCRGs in diseases, virus-infected cells, and pro-atherogenic DAMP-treated ECs; (7) OCRG expressions are significantly modulated in all the 28 cancer datasets, and the upregulated OCRGs are correlated with tumor immune infiltrates in some tumors; (8) tumor promoter factor IKK2 and tumor suppressor Tp53 significantly modulate the expressions of OCRGs. Our findings provide novel insights on the roles of upregulated OCRGs in the pathogenesis of inflammatory diseases and cancers, and novel pathways for the future therapeutic interventions for inflammations, sepsis, trauma, organ failures, autoimmune diseases, metabolic cardiovascular diseases (CVDs), and cancers.
Collapse
Affiliation(s)
- Ming Liu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, China
| | - Na Wu
- Departments of Endocrinology and Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Keman Xu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Fatma Saaoud
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Eleni Vasilopoulos
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ruijing Zhang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Jirong Wang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Cardiology, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Haitao Shen
- Departments of Endocrinology and Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | | | - Yifan Lu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yu Sun
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Charles Drummer
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Lu Liu
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Li Li
- Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, China
| | - Wenhui Hu
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jun Yu
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Domenico Praticò
- Alzheimer's Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jianxin Sun
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research, Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
143
|
Madyaningrana K, Vijayan V, Nikolin C, Aljabri A, Tumpara S, Korenbaum E, Shah H, Stankov M, Fuchs H, Janciauskiene S, Immenschuh S. Alpha1-antitrypsin counteracts heme-induced endothelial cell inflammatory activation, autophagy dysfunction and death. Redox Biol 2021; 46:102060. [PMID: 34246063 PMCID: PMC8274343 DOI: 10.1016/j.redox.2021.102060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 06/27/2021] [Indexed: 11/04/2022] Open
Abstract
Free heme toxicity in the vascular endothelium is critical for the pathogenesis of hemolytic disorders including sickle cell disease. In the current study, it is demonstrated that human alpha1-antitrypsin (A1AT), a serine protease inhibitor with high binding-affinity for heme, rescues endothelial cell (EC) injury caused by free heme. A1AT provided endothelial protection against free heme toxicity via a pathway that differs from human serum albumin and hemopexin, two prototypical heme-binding proteins. A1AT inhibited heme-mediated pro-inflammatory activation and death of ECs, but did not affect the increase in intracellular heme levels and up-regulation of the heme-inducible enzyme heme oxygenase-1. Moreover, A1AT reduced heme-mediated generation of mitochondrial reactive oxygen species. Extracellular free heme led to an increased up-take of A1AT by ECs, which was detected in lysosomes and was found to reduce heme-dependent alkalization of these organelles. Finally, A1AT was able to restore heme-dependent dysfunctional autophagy in ECs. Taken together, our findings show that A1AT rescues ECs from free heme-mediated pro-inflammatory activation, cell death and dysfunctional autophagy. Hence, A1AT therapy may be useful in the treatment of hemolytic disorders such as sickle cell disease.
Collapse
Affiliation(s)
- Kukuh Madyaningrana
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany; Faculty of Biotechnology, Universitas Kristen Duta Wacana, Yogyakarta, Indonesia
| | - Vijith Vijayan
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Christoph Nikolin
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Abid Aljabri
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Srinu Tumpara
- Department of Pulmonology, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Elena Korenbaum
- Institute for Biophysical Chemistry Hannover Medical School, Hannover, Germany
| | - Harshit Shah
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Metodi Stankov
- Department for Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Heiko Fuchs
- Institute of Experimental Ophthalmology, Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Pulmonology, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Stephan Immenschuh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
144
|
Chin MY, Patwardhan AR, Ang KH, Wang AL, Alquezar C, Welch M, Nguyen PT, Grabe M, Molofsky AV, Arkin MR, Kao AW. Genetically Encoded, pH-Sensitive mTFP1 Biosensor for Probing Lysosomal pH. ACS Sens 2021; 6:2168-2180. [PMID: 34102054 PMCID: PMC8240087 DOI: 10.1021/acssensors.0c02318] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/08/2021] [Indexed: 12/15/2022]
Abstract
Lysosomes are important sites for macromolecular degradation, defined by an acidic lumenal pH of ∼4.5. To better understand lysosomal pH, we designed a novel, genetically encoded, fluorescent protein (FP)-based pH biosensor called Fluorescence Indicator REporting pH in Lysosomes (FIRE-pHLy). This biosensor was targeted to lysosomes with lysosomal-associated membrane protein 1 (LAMP1) and reported lumenal pH between 3.5 and 6.0 with monomeric teal fluorescent protein 1 (mTFP1), a bright cyan pH-sensitive FP variant with a pKa of 4.3. Ratiometric quantification was enabled with cytosolically oriented mCherry using high-content quantitative imaging. We expressed FIRE-pHLy in several cellular models and quantified the alkalinizing response to bafilomycin A1, a specific V-ATPase inhibitor. In summary, we have engineered FIRE-pHLy, a specific, robust, and versatile lysosomal pH biosensor, that has broad applications for investigating pH dynamics in aging- and lysosome-related diseases, as well as in lysosome-based drug discovery.
Collapse
Affiliation(s)
- Marcus Y Chin
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Anand R Patwardhan
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Kean-Hooi Ang
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Austin L Wang
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Carolina Alquezar
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Mackenzie Welch
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Phi T Nguyen
- Weill Institute for Neurosciences, Department of Psychiatry, University of California, San Francisco, California 94158, United States
| | - Michael Grabe
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Anna V Molofsky
- Weill Institute for Neurosciences, Department of Psychiatry, University of California, San Francisco, California 94158, United States
| | - Michelle R Arkin
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Aimee W Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| |
Collapse
|
145
|
Lakpa KL, Khan N, Afghah Z, Chen X, Geiger JD. Lysosomal Stress Response (LSR): Physiological Importance and Pathological Relevance. J Neuroimmune Pharmacol 2021; 16:219-237. [PMID: 33751445 PMCID: PMC8099033 DOI: 10.1007/s11481-021-09990-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/08/2021] [Indexed: 02/08/2023]
Abstract
Extensive work has characterized endoplasmic reticulum (ER) and mitochondrial stress responses. In contrast, very little has been published about stress responses in lysosomes; subcellular acidic organelles that are physiologically important and are of pathological relevance. The greater lysosomal system is dynamic and is comprised of endosomes, lysosomes, multivesicular bodies, autophagosomes, and autophagolysosomes. They are important regulators of cellular physiology, they represent about 5% of the total cellular volume, they are heterogeneous in their sizes and distribution patterns, they are electron dense, and their subcellular positioning within cells varies in response to stimuli, insults and pH. These organelles are also integral to the pathogenesis of lysosomal storage diseases and it is increasingly recognized that lysosomes play important roles in the pathogenesis of such diverse conditions as neurodegenerative disorders and cancer. The purpose of this review is to focus attention on lysosomal stress responses (LSR), compare LSR with better characterized stress responses in ER and mitochondria, and form a framework for future characterizations of LSR. We synthesized data into the concept of LSR and present it here such that the definition of LSR can be modified as new knowledge is added and specific therapeutics are developed.
Collapse
Affiliation(s)
- Koffi L Lakpa
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Nabab Khan
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Zahra Afghah
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA
| | - Jonathan D Geiger
- Department of Biomedical Sciences, Dakota School of Medicine and Health Sciences, University of North, Grand Forks, ND, 58203, USA.
| |
Collapse
|
146
|
Chakrabarty RP, Chandel NS. Mitochondria as Signaling Organelles Control Mammalian Stem Cell Fate. Cell Stem Cell 2021; 28:394-408. [PMID: 33667360 DOI: 10.1016/j.stem.2021.02.011] [Citation(s) in RCA: 239] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent evidence supports the notion that mitochondrial metabolism is necessary for the determination of stem cell fate. Historically, mitochondrial metabolism is linked to the production of ATP and tricarboxylic acid (TCA) cycle metabolites to support stem cell survival and growth, respectively. However, it is now clear that beyond these canonical roles, mitochondria as signaling organelles dictate stem cell fate and function. In this review, we focus on key conceptual ideas on how mitochondria control mammalian stem cell fate and function through reactive oxygen species (ROS) generation, TCA cycle metabolite production, NAD+/NADH ratio regulation, pyruvate metabolism, and mitochondrial dynamics.
Collapse
Affiliation(s)
- Ram Prosad Chakrabarty
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Navdeep S Chandel
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
147
|
Basak I, Wicky HE, McDonald KO, Xu JB, Palmer JE, Best HL, Lefrancois S, Lee SY, Schoderboeck L, Hughes SM. A lysosomal enigma CLN5 and its significance in understanding neuronal ceroid lipofuscinosis. Cell Mol Life Sci 2021; 78:4735-4763. [PMID: 33792748 PMCID: PMC8195759 DOI: 10.1007/s00018-021-03813-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 01/09/2023]
Abstract
Neuronal Ceroid Lipofuscinosis (NCL), also known as Batten disease, is an incurable childhood brain disease. The thirteen forms of NCL are caused by mutations in thirteen CLN genes. Mutations in one CLN gene, CLN5, cause variant late-infantile NCL, with an age of onset between 4 and 7 years. The CLN5 protein is ubiquitously expressed in the majority of tissues studied and in the brain, CLN5 shows both neuronal and glial cell expression. Mutations in CLN5 are associated with the accumulation of autofluorescent storage material in lysosomes, the recycling units of the cell, in the brain and peripheral tissues. CLN5 resides in the lysosome and its function is still elusive. Initial studies suggested CLN5 was a transmembrane protein, which was later revealed to be processed into a soluble form. Multiple glycosylation sites have been reported, which may dictate its localisation and function. CLN5 interacts with several CLN proteins, and other lysosomal proteins, making it an important candidate to understand lysosomal biology. The existing knowledge on CLN5 biology stems from studies using several model organisms, including mice, sheep, cattle, dogs, social amoeba and cell cultures. Each model organism has its advantages and limitations, making it crucial to adopt a combinatorial approach, using both human cells and model organisms, to understand CLN5 pathologies and design drug therapies. In this comprehensive review, we have summarised and critiqued existing literature on CLN5 and have discussed the missing pieces of the puzzle that need to be addressed to develop an efficient therapy for CLN5 Batten disease.
Collapse
Affiliation(s)
- I Basak
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - H E Wicky
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - K O McDonald
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - J B Xu
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - J E Palmer
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - H L Best
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
- School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Wales, CF10 3AX, United Kingdom
| | - S Lefrancois
- Centre INRS-Institut Armand-Frappier, INRS, Laval, H7V 1B7, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, H3A 2B2, Canada
| | - S Y Lee
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - L Schoderboeck
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - S M Hughes
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand.
| |
Collapse
|
148
|
Mochizuki-Kashio M, Shiozaki H, Suda T, Nakamura-Ishizu A. Mitochondria Turnover and Lysosomal Function in Hematopoietic Stem Cell Metabolism. Int J Mol Sci 2021; 22:4627. [PMID: 33924874 PMCID: PMC8124492 DOI: 10.3390/ijms22094627] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 01/17/2023] Open
Abstract
Hematopoietic stem cells (HSCs) reside in a hypoxic microenvironment that enables glycolysis-fueled metabolism and reduces oxidative stress. Nonetheless, metabolic regulation in organelles such as the mitochondria and lysosomes as well as autophagic processes have been implicated as essential for the determination of HSC cell fate. This review encompasses the current understanding of anaerobic metabolism in HSCs as well as the emerging roles of mitochondrial metabolism and lysosomal regulation for hematopoietic homeostasis.
Collapse
Affiliation(s)
- Makiko Mochizuki-Kashio
- Microanatomy and Developmental Biology, Tokyo Women’s Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan;
| | - Hiroko Shiozaki
- Department of Hematology, Tokyo Women’s Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan;
| | - Toshio Suda
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, Singapore 117599, Singapore;
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan
| | - Ayako Nakamura-Ishizu
- Microanatomy and Developmental Biology, Tokyo Women’s Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan;
| |
Collapse
|
149
|
Ramos VDM, Kowaltowski AJ, Kakimoto PA. Autophagy in Hepatic Steatosis: A Structured Review. Front Cell Dev Biol 2021; 9:657389. [PMID: 33937257 PMCID: PMC8081956 DOI: 10.3389/fcell.2021.657389] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/16/2021] [Indexed: 01/18/2023] Open
Abstract
Steatosis is the accumulation of neutral lipids in the cytoplasm. In the liver, it is associated with overeating and a sedentary lifestyle, but may also be a result of xenobiotic toxicity and genetics. Non-alcoholic fatty liver disease (NAFLD) defines an array of liver conditions varying from simple steatosis to inflammation and fibrosis. Over the last years, autophagic processes have been shown to be directly associated with the development and progression of these conditions. However, the precise role of autophagy in steatosis development is still unclear. Specifically, autophagy is necessary for the regulation of basic metabolism in hepatocytes, such as glycogenolysis and gluconeogenesis, response to insulin and glucagon signaling, and cellular responses to free amino acid contents. Also, genetic knockout models for autophagy-related proteins suggest a critical relationship between autophagy and hepatic lipid metabolism, but some results are still ambiguous. While autophagy may seem necessary to support lipid oxidation in some contexts, other evidence suggests that autophagic activity can lead to lipid accumulation instead. This structured literature review aims to critically discuss, compare, and organize results over the last 10 years regarding rodent steatosis models that measured several autophagy markers, with genetic and pharmacological interventions that may help elucidate the molecular mechanisms involved.
Collapse
Affiliation(s)
| | | | - Pamela A. Kakimoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
150
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Puebla A, Lovy A, Cesar Cardenas J. The ER-mitochondria Ca 2+ signaling in cancer progression: Fueling the monster. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:49-121. [PMID: 34392932 DOI: 10.1016/bs.ircmb.2021.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a leading cause of death worldwide. All major tumor suppressors and oncogenes are now recognized to have fundamental connections with metabolic pathways. A hallmark feature of cancer cells is a reprogramming of their metabolism even when nutrients are available. Increasing evidence indicates that most cancer cells rely on mitochondrial metabolism to sustain their energetic and biosynthetic demands. Mitochondria are functionally and physically coupled to the endoplasmic reticulum (ER), the major calcium (Ca2+) storage organelle in mammalian cells, through special domains known as mitochondria-ER contact sites (MERCS). In this domain, the release of Ca2+ from the ER is mainly regulated by inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), a family of Ca2+ release channels activated by the ligand IP3. IP3R mediated Ca2+ release is transferred to mitochondria through the mitochondrial Ca2+ uniporter (MCU). Once in the mitochondrial matrix, Ca2+ activates several proteins that stimulate mitochondrial performance. The role of IP3R and MCU in cancer, as well as the other proteins that enable the Ca2+ communication between these two organelles is just beginning to be understood. Here, we describe the function of the main players of the ER mitochondrial Ca2+ communication and discuss how this particular signal may contribute to the rise and development of cancer traits.
Collapse
Affiliation(s)
- Galdo Bustos
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Eduardo Silva-Pavez
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Andrea Puebla
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Alenka Lovy
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Department of Neuroscience, Center for Neuroscience Research, Tufts School of Medicine, Boston, MA, United States.
| | - J Cesar Cardenas
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, United States; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States.
| |
Collapse
|