1
|
McKay M, Gorai S, Paidi RK, Mondal S, Pahan K. Identification of Cinnamein, a Component of Balsam of Tolu/Peru, as a New Ligand of PPARα for Plaque Reduction and Memory Protection in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis Rep 2024; 8:903-922. [PMID: 38910936 PMCID: PMC11191634 DOI: 10.3233/adr-230179] [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: 12/06/2023] [Accepted: 04/15/2024] [Indexed: 06/25/2024] Open
Abstract
Background Despite intense investigations, no effective treatment is yet available to reduce plaques and protect memory and learning in patients with Alzheimer's disease (AD), the most common neurodegenerative disorder. Therefore, it is important to identify a non-toxic, but effective, treatment option for AD. Objective Cinnamein, a nontoxic compound, is naturally available in Balsam of Peru and Tolu Balsam. We examined whether cinnamein treatment could decrease plaques and improve cognitive functions in 5XFAD mouse model of AD. Methods We employed in silico analysis, time-resolved fluorescence energy transfer assay, thermal shift assay, primary neuron isolation, western blot, immunostaining, immunohistochemistry, Barnes maze, T maze, and open field behavior. Results Oral administration of cinnamein led to significant reduction in amyloid-β plaque deposits in the brain and protection of spatial learning and memory in 5XFAD mice. Peroxisome proliferator-activated receptor alpha (PPARα), a nuclear hormone receptor, is involved in plaque lowering and increase in hippocampal plasticity. While investigating underlying mechanisms, we found that cinnamein served as a ligand of PPARα. Accordingly, oral cinnamein upregulated the level of PPARα, but not PPARβ, in the hippocampus, and remained unable to decrease plaques from the hippocampus and improve memory and learning in 5XFAD mice lacking PPARα. While A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) is one of the drivers of nonamyloidogenic pathway, transcription factor EB (TFEB) is considered as the master regulator of autophagy. Cinnamein treatment was found to upregulate both ADAM10 and TFEB in the brain of 5XFAD mice via PPARα. Conclusions Our results suggest that this balsam component may have therapeutic importance in AD.
Collapse
Affiliation(s)
- Mary McKay
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Ramesh K. Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| |
Collapse
|
2
|
Raha S, Paidi RK, Dutta D, Pahan K. Cinnamic acid, a natural plant compound, exhibits neuroprotection in a mouse model of Sandhoff disease via PPARα. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:17-32. [PMID: 38532783 PMCID: PMC10961485 DOI: 10.1515/nipt-2023-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Tay-Sachs disease (TSD) and its severe form Sandhoff disease (SD) are autosomal recessive lysosomal storage metabolic disorders, which often result into excessive GM2 ganglioside accumulation predominantly in lysosomes of nerve cells. Although patients with these diseases appear normal at birth, the progressive accumulation of undegraded GM2 gangliosides in neurons leads to early death accompanied by manifestation of motor difficulties and gradual loss of behavioral skills. Unfortunately, there is still no effective treatment available for TSD/SD. The present study highlights the importance of cinnamic acid (CA), a naturally occurring aromatic fatty acid present in a number of plants, in inhibiting the disease process in a transgenic mouse model of SD. Oral administration of CA significantly attenuated glial activation and inflammation and reduced the accumulation of GM2 gangliosides/glycoconjugates in the cerebral cortex of Sandhoff mice. Besides, oral CA also improved behavioral performance and increased the survival of Sandhoff mice. While assessing the mechanism, we found that oral administration of CA increased the level of peroxisome proliferator-activated receptor α (PPARα) in the brain of Sandhoff mice and that oral CA remained unable to reduce glycoconjugates, improve behavior and increase survival in Sandhoff mice lacking PPARα. Our results indicate a beneficial function of CA that utilizes a PPARα-dependent mechanism to halt the progression of SD and thereby increase the longevity of Sandhoff mice.
Collapse
Affiliation(s)
- Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Ramesh K. Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
3
|
Paidi RK, Raha S, Roy A, Pahan K. Muscle-building supplement β-hydroxy β-methylbutyrate binds to PPARα to improve hippocampal functions in mice. Cell Rep 2023; 42:112717. [PMID: 37437568 PMCID: PMC10440158 DOI: 10.1016/j.celrep.2023.112717] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 04/09/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023] Open
Abstract
This study underlines the importance of β-hydroxy β-methylbutyrate (HMB), a muscle-building supplement in human, in increasing mouse hippocampal plasticity. Detailed proteomic analyses reveal that HMB serves as a ligand of peroxisome proliferator-activated receptor α (PPARα), a nuclear hormone receptor involved in fat metabolism, via interaction with the Y314 residue. Accordingly, HMB is ineffective in increasing plasticity of PPARα-/- hippocampal neurons. While lentiviral establishment of full-length PPARα restores the plasticity-promoting effect of HMB in PPARα-/- hippocampal neurons, lentiviral transduction of Y314D-PPARα remains unable to do that, highlighting the importance of HMB's interaction with the Y314 residue. Additionally, oral HMB improves spatial learning and memory and reduces plaque load in 5X familial Alzheimer's disease (5XFAD) mice, but not in 5XFADΔPPARα mice (5XFAD lacking PPARα), indicating the involvement of PPARα in HMB-mediated neuroprotection in 5XFAD mice. These results delineate neuroprotective functions of HMB and suggest that this widely used supplement may be repurposed for AD.
Collapse
Affiliation(s)
- Ramesh K Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Avik Roy
- Simmaron Research Institute, Technology Innovation Center, 10437 W Innovation Drive, Wauwatosa, WI, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| |
Collapse
|
4
|
Jana M, Dutta D, Poddar J, Pahan K. Activation of PPARα Exhibits Therapeutic Efficacy in a Mouse Model of Juvenile Neuronal Ceroid Lipofuscinosis. J Neurosci 2023; 43:1814-1829. [PMID: 36697260 PMCID: PMC10010460 DOI: 10.1523/jneurosci.2447-21.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Juvenile neuronal ceroid lipofuscinosis (JNCL) is a fatal inherited neurodegenerative disease of children that occurs because of defective function of the lysosomal membrane glycoprotein CLN3. JNCL features glial activation and accumulation of autofluorescent storage material containing subunit c of mitochondrial ATP synthase (SCMAS), ultimately resulting into neuronal loss. Until now, no effective therapy is available for JNCL. This study underlines the possible therapeutic importance of gemfibrozil, an activator of peroxisome proliferator-activated receptor α (PPARα) and a lipid-lowering drug approved by the Food and Drug Administration in an animal model of JNCL. Oral gemfibrozil treatment reduced microglial and astroglial activation, attenuated neuroinflammation, restored the level of transcription factor EB (TFEB; the master regulator of lysosomal biogenesis), and decreased the accumulation of storage material SCMAS in somatosensory barrel field (SBF) cortex of Cln3Δex7/8 (Cln3ΔJNCL) mice of both sexes. Accordingly, gemfibrozil treatment also improved locomotor activities of Cln3ΔJNCL mice. While investigating the mechanism, we found marked loss of PPARα in the SBF cortex of Cln3ΔJNCL mice, which increased after gemfibrozil treatment. Oral gemfibrozil also stimulated the recruitment of PPARα to the Tfeb gene promoter in vivo in the SBF cortex of Cln3ΔJNCL mice, indicating increased transcription of Tfeb in the CNS by gemfibrozil treatment via PPARα. Moreover, disease pathologies aggravated in Cln3ΔJNCL mice lacking PPARα (Cln3ΔJNCLΔPPARα) and gemfibrozil remained unable to decrease SCMAS accumulation, reduce glial activation, and improve locomotor performance of Cln3ΔJNCLΔPPARα mice. These results suggest that activation of PPARα may be beneficial for JNCL and that gemfibrozil may be repurposed for the treatment of this incurable disease.SIGNIFICANCE STATEMENT Despite intense investigations, no effective therapy is available for JNCL, an incurable inherited lysosomal storage disorder. Here, we delineate that oral administration of gemfibrozil, a lipid-lowering drug, decreases glial inflammation, normalizes and/or upregulates TFEB, and reduces accumulation of autofluorescent storage material in SBF cortex to improve locomotor activities in Cln3Δex7/8 (Cln3ΔJNCL) mice. Aggravation of disease pathology in Cln3ΔJNCL mice lacking PPARα (Cln3ΔJNCLΔPPARα) and inability of gemfibrozil to decrease SCMAS accumulation, reduce glial activation, and improve locomotor performance of Cln3ΔJNCLΔPPARα mice delineates an important role of PPARα in this process. These studies highlight a new property of gemfibrozil and indicate its possible therapeutic use in JNCL patients.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
| | - Jit Poddar
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| |
Collapse
|
5
|
Dutta D, Paidi RK, Raha S, Roy A, Chandra S, Pahan K. Treadmill exercise reduces α-synuclein spreading via PPARα. Cell Rep 2022; 40:111058. [PMID: 35830804 PMCID: PMC9308946 DOI: 10.1016/j.celrep.2022.111058] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/29/2021] [Accepted: 06/15/2022] [Indexed: 11/30/2022] Open
Abstract
This study underlines the importance of treadmill exercise in reducing α-synuclein (α-syn) spreading in the A53T brain and protecting nigral dopaminergic neurons. Preformed α-syn fibril (PFF) seeding in the internal capsule of young A53T α-syn mice leads to increased spreading of α-syn to substantia nigra and motor cortex and concomitant loss of nigral dopaminergic neurons. However, regular treadmill exercise decreases α-syn spreading in the brain and protects nigral dopaminergic neurons in PFF-seeded mice. Accordingly, treadmill exercise also mitigates α-synucleinopathy in aged A53T mice. While investigating this mechanism, we have observed that treadmill exercise induces the activation of peroxisome proliferator-activated receptor α (PPARα) in the brain to stimulate lysosomal biogenesis via TFEB. Accordingly, treadmill exercise remains unable to stimulate TFEB and reduce α-synucleinopathy in A53T mice lacking PPARα, and fenofibrate, a prototype PPARα agonist, reduces α-synucleinopathy. These results delineate a beneficial function of treadmill exercise in reducing α-syn spreading in the brain via PPARα.
Collapse
Affiliation(s)
- Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ramesh Kumar Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Sujyoti Chandra
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| |
Collapse
|
6
|
Hao Y, Su C, Liu X, Sui H, Shi Y, Zhao L. Bioengineered microglia-targeted exosomes facilitate Aβ clearance via enhancing activity of microglial lysosome for promoting cognitive recovery in Alzheimer's disease. BIOMATERIALS ADVANCES 2022; 136:212770. [PMID: 35929330 DOI: 10.1016/j.bioadv.2022.212770] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/08/2022] [Accepted: 03/17/2022] [Indexed: 06/15/2023]
Abstract
Aggregation of amyloid in the form of senile plaques is currently considered to be one of the main mechanisms driving the development of Alzheimer's disease (AD). Therefore, targeting amyloid homeostasis is an important treatment strategy for AD. Microglia, as the main immune cells, contribute to endocytosis and clearance of amyloid beta (Aβ) via lysosome mediated degradation. As abnormal lysosomal function in microglia is associated with inefficient clearance of Aβ in AD, we designed bioengineered microglia-targeting exosomes to promote the targeted delivery of gemfibrozil (Gem) and restore the lysosomal activity of microglia in clearing Aβ aggregation. Our results suggested that mannose-modified exosomes laden with Gem (MExo-Gem) can not only bind with Aβ but also specifically target microglia through the interaction between Exo-delivered mannose and mannose receptors expressed in microglia, thus promoting Aβ entry into microglia. Exosomal Gem activated lysosomal activity and accelerated lysosome-mediated clearance of Aβ in microglia. Finally, MExo-Gem improved the learning and memory ability of AD model mice.
Collapse
Affiliation(s)
- Yunni Hao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Chang Su
- School of Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Xintong Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Haijuan Sui
- Department of Pharmacology, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| |
Collapse
|
7
|
Iyaswamy A, Krishnamoorthi SK, Zhang H, Sreenivasmurthy SG, Zhu Z, Liu J, Su CF, Guan XJ, Wang ZY, Cheung KH, Song JX, Durairajan SSK, Li M. Qingyangshen mitigates amyloid-β and Tau aggregate defects involving PPARα-TFEB activation in transgenic mice of Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153648. [PMID: 34332287 DOI: 10.1016/j.phymed.2021.153648] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/01/2021] [Accepted: 06/30/2021] [Indexed: 05/26/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease. Deposition of amyloid β plaques (Aβ) and neurofibrillary tangles (NFTs) is the key pathological hallmark of AD. Accumulating evidence suggest that impairment of autophagy-lysosomal pathway (ALP) plays key roles in AD pathology. PURPOSE The present study aims to assess the neuroprotective effects of Qingyangshen (QYS), a Chinese herbal medicine, in AD cellular and animal models and to determine its underlying mechanisms involving ALP regulation. METHODS QYS extract was prepared and its chemical components were characterized by LC/MS. Then the pharmacokinetics and acute toxicity of QYS extract were evaluated. The neuroprotective effects of QYS extract were determined in 3XTg AD mice, by using a series of behavioral tests and biochemical assays, and the mechanisms were examined in vitro. RESULTS Oral administration of QYS extract improved learning and spatial memory, reduced carboxy-terminal fragments (CTFs), amyloid precursor protein (APP), Aβ and Tau aggregates, and inhibited microgliosis and astrocytosis in the brains of 3XTg mice. Mechanistically, QYS extract increased the expression of PPARα and TFEB, and promoted ALP both in vivo and in vitro. CONCLUSION QYS attenuates AD pathology, and improves cognitive function in 3XTg mice, which may be mediated by activation of PPARα-TFEB pathway and the subsequent ALP enhancement. Therefore, QYS may be a promising herbal material for further anti-AD drug discovery.
Collapse
Affiliation(s)
- Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Senthil Kumar Krishnamoorthi
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Huan Zhang
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sravan G Sreenivasmurthy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zhou Zhu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jia Liu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Cheng-Fu Su
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xin-Jie Guan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zi-Ying Wang
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, China
| | - King-Ho Cheung
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ju-Xian Song
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Siva Sundara Kumar Durairajan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Division of Mycobiology and Neurodegenerative disease research, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India.
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
8
|
Yoo J, Jeong IK, Ahn KJ, Chung HY, Hwang YC. Fenofibrate, a PPARα agonist, reduces hepatic fat accumulation through the upregulation of TFEB-mediated lipophagy. Metabolism 2021; 120:154798. [PMID: 33984335 DOI: 10.1016/j.metabol.2021.154798] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/18/2021] [Accepted: 05/09/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND Recent studies have shown that dysregulation of autophagy is involved in the development of nonalcoholic fatty liver disease (NAFLD). Transcription factors E3 (TFE3) and EB (TFEB) are master regulators of the transcriptional response of basic cellular processes such as lysosomal biogenesis and autophagy. Here, we investigated the role of fenofibrate, a PPARα agonist, in promotion of intracellular lipid clearance by upregulation of TFEB/TFE3. METHODS We investigated whether the effects of fenofibrate on livers were dependent on TFEB in high fat diet (HFD)-fed mice and in vivo Tfeb knockdown mice. These mice were analyzed for characteristics of obesity and diabetes; the effects of fenofibrate on hepatic fat content, glucose sensitivity, insulin resistance, and autophagy functional dependence on TFEB were investigated. HepG2, Hep3B, TSC2+/+ and tsc2-/- MEFs, tfeb wild type- and tfeb knockout-HeLa cells were used for in vitro experiments. RESULTS Fenofibrate treatment activated autophagy and TFEB/TFE3 and reduced hepatic fat accumulation in an mTOR-independent manner. Knockdown of TFEB offset the effects of fenofibrate on autophagy and hepatic fat accumulation. In addition, fenofibrate treatment induced lysosomal Ca2+ release through mucolipin 1, activated calcineurin and the CaMKKβ-AMPK-ULK1 pathway, subsequently promoted TFEB and TFE3 dephosphorylation and nuclear translocation. Treatment with calcium chelator or knockdown of mucolipin 1 in hepatocytes offset the effects of fenofibrate treatment on autophagy and hepatic fat accumulation. CONCLUSION Activation of PPARα ameliorates hepatic fat accumulation via activation of TFEB and lipophagy induction. Lysosomal calcium signaling appears to play a critical role in this process. In addition, activation of TFEB by modulating nuclear receptors including PPARα with currently available drugs or new molecules might be a therapeutic target for treatment of NAFLD and other cardiometabolic diseases.
Collapse
Affiliation(s)
- Jin Yoo
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Endocrinology and Metabolism, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - In-Kyung Jeong
- Department of Endocrinology and Metabolism, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Kyu Jeung Ahn
- Department of Endocrinology and Metabolism, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Ho Yeon Chung
- Department of Endocrinology and Metabolism, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - You-Cheol Hwang
- Department of Endocrinology and Metabolism, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Tao H, Yancey PG, Blakemore JL, Zhang Y, Ding L, Jerome WG, Brown JD, Vickers KC, Linton MF. Macrophage SR-BI modulates autophagy via VPS34 complex and PPARα transcription of Tfeb in atherosclerosis. J Clin Invest 2021; 131:94229. [PMID: 33661763 PMCID: PMC8011903 DOI: 10.1172/jci94229] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Autophagy modulates lipid turnover, cell survival, inflammation, and atherogenesis. Scavenger receptor class B type I (SR-BI) plays a crucial role in lysosome function. Here, we demonstrate that SR-BI regulates autophagy in atherosclerosis. SR-BI deletion attenuated lipid-induced expression of autophagy mediators in macrophages and atherosclerotic aortas. Consequently, SR-BI deletion resulted in 1.8- and 2.5-fold increases in foam cell formation and apoptosis, respectively, and increased oxidized LDL-induced inflammatory cytokine expression. Pharmacological activation of autophagy failed to reduce lipid content or apoptosis in Sr-b1-/- macrophages. SR-BI deletion reduced both basal and inducible levels of transcription factor EB (TFEB), a master regulator of autophagy, causing decreased expression of autophagy genes encoding VPS34 and Beclin-1. Notably, SR-BI regulated Tfeb expression by enhancing PPARα activation. Moreover, intracellular macrophage SR-BI localized to autophagosomes, where it formed cholesterol domains resulting in enhanced association of Barkor and recruitment of the VPS34-Beclin-1 complex. Thus, SR-BI deficiency led to lower VPS34 activity in macrophages and in atherosclerotic aortic tissues. Overexpression of Tfeb or Vps34 rescued the defective autophagy in Sr-b1-/- macrophages. Taken together, our results show that macrophage SR-BI regulates autophagy via Tfeb expression and recruitment of the VPS34-Beclin-1 complex, thus identifying previously unrecognized roles for SR-BI and potentially novel targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Huan Tao
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Patricia G. Yancey
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - John L. Blakemore
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Youmin Zhang
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Lei Ding
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - W. Gray Jerome
- Department of Pathology, Microbiology and Immunology, and
| | - Jonathan D. Brown
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - Kasey C. Vickers
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
| | - MacRae F. Linton
- Department of Medicine, Atherosclerosis Research Unit, Division of Cardiovascular Medicine
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
10
|
Morris R, Armbruster K, Silva J, Widell DJ, Cheng F. The Association between the Usage of Non-Steroidal Anti-Inflammatory Drugs and Cognitive Status: Analysis of Longitudinal and Cross-Sectional Studies from the Global Alzheimer's Association Interactive Network and Transcriptomic Data. Brain Sci 2020; 10:brainsci10120961. [PMID: 33321871 PMCID: PMC7763310 DOI: 10.3390/brainsci10120961] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 01/09/2023] Open
Abstract
The degenerative cognitive and physical decline of Alzheimer patients, coupled with the extensive psychological and economic tolls imposed on family members that serve as caretakers, necessitate the discovery of effective cures and preventative measures for age-related cognitive depreciation. In the journey of Alzheimer’s disease treatment discovery, several cross-sectional and longitudinal studies have delineated a noticeable association between the use of nonsteroidal anti-inflammatory drugs (NSAIDs), a class of low-cost drugs with minimal side effects, and the alleviation of age-related memory impairment. In this study, four datasets (two cross-sectional and two longitudinal studies) derived from the Global Alzheimer’s Association Interactive Network (GAAIN) were analyzed. The significant association between the usage of NSAIDs and better cognitive status was observed. The results agree with the findings of previous studies that the use of NSAIDs may be beneficial in the early stages of Alzheimer’s disease. Transcriptomic data show that ibuprofen treatment results in upregulation of several genes involved in arachidonic acid metabolism including PPARγ, Cyp4a12b, Cyp2c66, and Cyp2c37 in the hippocampus. The increase in conversion of arachidonic acid into anti-inflammatory 16C and 18C dicarboxylic acids as well as epoxyeicosatrienoic acids may play a role in reducing the risk of Alzheimer’s disease development.
Collapse
Affiliation(s)
- Robert Morris
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL 33613, USA;
| | - Kyle Armbruster
- Biomedical Science Program, College of Art and Science, University of South Florida, Tampa, FL 33620, USA; (K.A.); (J.S.); (D.J.W.)
| | - Julianna Silva
- Biomedical Science Program, College of Art and Science, University of South Florida, Tampa, FL 33620, USA; (K.A.); (J.S.); (D.J.W.)
| | - Daniel James Widell
- Biomedical Science Program, College of Art and Science, University of South Florida, Tampa, FL 33620, USA; (K.A.); (J.S.); (D.J.W.)
| | - Feng Cheng
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL 33613, USA;
- Correspondence: ; Tel.: +1-813-974-4288
| |
Collapse
|
11
|
Paik S, Jo EK. An Interplay Between Autophagy and Immunometabolism for Host Defense Against Mycobacterial Infection. Front Immunol 2020; 11:603951. [PMID: 33262773 PMCID: PMC7688515 DOI: 10.3389/fimmu.2020.603951] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/16/2020] [Indexed: 12/15/2022] Open
Abstract
Autophagy, an intracellular catabolic pathway featuring lysosomal degradation, is a central component of the host immune defense against various infections including Mycobacterium tuberculosis (Mtb), the pathogen that causes tuberculosis. Mtb can evade the autophagic defense and drive immunometabolic remodeling of host phagocytes. Co-regulation of the autophagic and metabolic pathways may play a pivotal role in shaping the innate immune defense and inflammation during Mtb infection. Two principal metabolic sensors, AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) kinase, function together to control the autophagy and immunometabolism that coordinate the anti-mycobacterial immune defense. Here, we discuss our current understanding of the interplay between autophagy and immunometabolism in terms of combating intracellular Mtb, and how AMPK-mTOR signaling regulates antibacterial autophagy in terms of Mtb infection. We describe several autophagy-targeting agents that promote host antimicrobial defenses by regulating the AMPK-mTOR axis. A better understanding of the crosstalk between immunometabolism and autophagy, both of which are involved in host defense, is crucial for the development of innovative targeted therapies for tuberculosis.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
12
|
Abstract
Mounting evidence has identified that impaired amyloid-β (Aβ) clearance might contribute to Alzheimer's disease (AD) pathology. The lysosome-autophagy network plays an important role in protein homeostasis and cell health by removing abnormal protein aggregates via intracellular degradation. Therefore, stimulation of cellular degradative machinery for efficient removal of Aβ has emerged as a growing field in AD research. However, mechanisms controlling such pathways and drugs to promote such mechanisms are poorly understood. Aspirin is a widely used drug throughout the world and recent studies have identified a new function of this drug. At low doses, aspirin stimulates lysosomal biogenesis and autophagy to clear amyloid plaques in an animal model of AD. This review delineates such functions of aspirin and analyzes underlying mechanisms that involve peroxisome proliferator-activated receptor alpha (PPARα)-mediated transcription of transcription factor EB (TFEB), the master regulator of lysosomal biogenesis.
Collapse
Affiliation(s)
- Sujyoti Chandra
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Dhruv R Patel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
13
|
Gomaraschi M, Fracanzani AL, Dongiovanni P, Pavanello C, Giorgio E, Da Dalt L, Norata GD, Calabresi L, Consonni D, Lombardi R, Branchi A, Fargion S. Lipid accumulation impairs lysosomal acid lipase activity in hepatocytes: Evidence in NAFLD patients and cell cultures. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:158523. [DOI: 10.1016/j.bbalip.2019.158523] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/27/2019] [Accepted: 09/05/2019] [Indexed: 12/23/2022]
|
14
|
Potential Effects of Aspirin on Lysosomal Biogenesis and Amyloid-β Clearance: An Old Drug and Novel Insights in Alzheimer's Disease Therapy. J Neurosci 2019; 39:197-198. [PMID: 30626724 DOI: 10.1523/jneurosci.2283-18.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/02/2018] [Accepted: 11/14/2018] [Indexed: 12/15/2022] Open
|
15
|
Pavanello C, Baragetti A, Branchi A, Grigore L, Castelnuovo S, Giorgio E, Catapano AL, Calabresi L, Gomaraschi M. Treatment with fibrates is associated with higher LAL activity in dyslipidemic patients. Pharmacol Res 2019; 147:104362. [PMID: 31330253 DOI: 10.1016/j.phrs.2019.104362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/01/2019] [Accepted: 07/18/2019] [Indexed: 01/01/2023]
Abstract
Lysosomal acid lipase (LAL) is responsible for the hydrolysis of cholesteryl esters (CE) and triglycerides (TG) within the lysosomes; generated cholesterol and free fatty acids (FFA) are released in the cytosol where they can regulate their own synthesis and metabolism. When LAL is not active, as in case of genetic mutations, CE and TG accumulate in the lysosomal compartment, while the lack of release of cholesterol and FFA in the cytosol leads to an upregulation of their synthesis. Thus, LAL plays a central role in the intracellular homeostasis of lipids. Since there are no indications about the effect of different lipid-lowering agents on LAL activity, aim of the study was to address the relationship between LAL activity and the type of lipid-lowering therapy in a cohort of dyslipidemic patients. LAL activity was measured on dried blood spot from 120 patients with hypercholesterolemia or mixed dyslipidemia and was negatively correlated to LDL-cholesterol levels. Among enrolled patients, ninety-one were taking one or more lipid-lowering drugs, as statins, fibrates, ezetimibe and omega-3 polyunsaturated fatty acids. When patients were stratified according to the type of lipid-lowering treatment, i.e. untreated, taking statins or taking fibrates, LAL activity was significantly higher in those with fibrates, even after adjustment for sex, age, BMI, lipid parameters, liver function, metabolic syndrome, diabetes and statin use. In a subset of patients tested after 3 months of treatment with micronized fenofibrate, LAL activity raised by 21%; the increase was negatively correlated with baseline LAL activity. Thus, the use of fibrates is independently associated with higher LAL activity in dyslipidemic patients, suggesting that the positive effects of PPAR-α activation on cellular and systemic lipid homeostasis can also include an improved LAL activity.
Collapse
Affiliation(s)
- Chiara Pavanello
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Andrea Baragetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; S.I.S.A. Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Adriana Branchi
- Centro per lo Studio e la Prevenzione dell'Aterosclerosi, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Milan, Italy
| | - Liliana Grigore
- S.I.S.A. Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Samuela Castelnuovo
- Centro Dislipidemie, Dipartimento Cardiotoracovascolare, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Eleonora Giorgio
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Alberico L Catapano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; IRCCS Multimedica Hospital, Milan, Italy
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Monica Gomaraschi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
16
|
Watch What You (Self-) Eat: Autophagic Mechanisms that Modulate Metabolism. Cell Metab 2019; 29:803-826. [PMID: 30943392 PMCID: PMC6450419 DOI: 10.1016/j.cmet.2019.03.003] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/07/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Abstract
Autophagy is an evolutionarily conserved lysosome- or vacuole-dependent catabolic pathway in eukaryotes. Autophagy functions basally for cellular quality control and is induced to act as an alternative source of basic metabolites during nutrient deprivation. These functions of autophagy are intimately connected to the regulation of metabolism, and the metabolic status of the cell in turn controls the nature and extent of autophagic induction. Here, we highlight the co-regulation of autophagy and metabolism with a special focus on selective autophagy that, along with bulk autophagy, plays a central role in regulating and rewiring metabolic circuits. We outline the metabolic signals that activate these pathways, the mechanisms involved, and the downstream effects and implications while recognizing yet unanswered questions. We also discuss the role of autophagy in the development and maintenance of adipose tissue, an emerging player in systemic metabolic homeostasis, and describe what is currently known about the complex relationship between autophagy and cancer.
Collapse
|
17
|
Prorok T, Jana M, Patel D, Pahan K. Cinnamic Acid Protects the Nigrostriatum in a Mouse Model of Parkinson's Disease via Peroxisome Proliferator-Activated Receptorα. Neurochem Res 2019; 44:751-762. [PMID: 30612307 DOI: 10.1007/s11064-018-02705-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/20/2018] [Accepted: 12/20/2018] [Indexed: 12/23/2022]
Abstract
Parkinson's disease (PD) is the second most common devastating human neurodegenerative disorder and despite intense investigation, no effective therapy is available for PD. Cinnamic acid, a naturally occurring aromatic fatty acid of low toxicity, is a precursor for the synthesis of a huge number of plant substances. This study highlights the neuroprotective effect of cinnamic acid in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Oral administration of cinnamic acid protected tyrosine hydroxylase (TH)-positive dopaminergic neurons in the substantia nigra pars compacta (SNpc) and TH fibers in the striatum of MPTP-insulted mice. Accordingly, oral cinnamic acid also normalized striatal neurotransmitters and improved locomotor activities in MPTP-intoxicated mice. While investigating mechanisms, we found that cinnamic acid induced the activation of peroxisome proliferator-activated receptor α (PPARα), but not PPARβ, in primary mouse astrocytes. Cinnamic acid mediated protection of the nigrostriatal system and locomotor activities in WT and PPARβ (-/-), but not PPARα (-/-) mice from MPTP intoxication suggests that cinnamic acid requires the involvement of PPARα in protecting dopaminergic neurons in this model of PD. This study delineates a new function of cinnamic acid in protecting dopaminergic neurons via PPARα that could be beneficial for PD.
Collapse
Affiliation(s)
- Tim Prorok
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA.,Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Malabendu Jana
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA.,Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Dhruv Patel
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA.,Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA. .,Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA.
| |
Collapse
|