85 results on '"Dhanendra Tomar"'
Search Results
2. Post-translational modifications and protein quality control of mitochondrial channels and transporters
- Author
-
Ashlesha Kadam, Pooja Jadiya, and Dhanendra Tomar
- Subjects
mitochondrial transporters ,mitochondrial channels ,posttranslational modifications ,MPQC ,MCU ,VDAC ,Biology (General) ,QH301-705.5 - Abstract
Mitochondria play a critical role in energy metabolism and signal transduction, which is tightly regulated by proteins, metabolites, and ion fluxes. Metabolites and ion homeostasis are mainly mediated by channels and transporters present on mitochondrial membranes. Mitochondria comprise two distinct compartments, the outer mitochondrial membrane (OMM) and the inner mitochondrial membrane (IMM), which have differing permeabilities to ions and metabolites. The OMM is semipermeable due to the presence of non-selective molecular pores, while the IMM is highly selective and impermeable due to the presence of specialized channels and transporters which regulate ion and metabolite fluxes. These channels and transporters are modulated by various post-translational modifications (PTMs), including phosphorylation, oxidative modifications, ions, and metabolites binding, glycosylation, acetylation, and others. Additionally, the mitochondrial protein quality control (MPQC) system plays a crucial role in ensuring efficient molecular flux through the mitochondrial membranes by selectively removing mistargeted or defective proteins. Inefficient functioning of the transporters and channels in mitochondria can disrupt cellular homeostasis, leading to the onset of various pathological conditions. In this review, we provide a comprehensive overview of the current understanding of mitochondrial channels and transporters in terms of their functions, PTMs, and quality control mechanisms.
- Published
- 2023
- Full Text
- View/download PDF
3. Neuronal loss of NCLX-dependent mitochondrial calcium efflux mediates age-associated cognitive decline
- Author
-
Pooja Jadiya, Henry M. Cohen, Devin W. Kolmetzky, Ashlesha A. Kadam, Dhanendra Tomar, and John W. Elrod
- Subjects
Behavioral neuroscience ,Molecular neuroscience ,Cellular neuroscience ,Cognitive neuroscience ,Science - Abstract
Summary: Mitochondrial calcium overload contributes to neurodegenerative disease development and progression. We recently reported that loss of the mitochondrial sodium/calcium exchanger (NCLX), the primary mechanism of mCa2+ efflux, promotes mCa2+ overload, metabolic derangement, redox stress, and cognitive decline in models of Alzheimer’s disease (AD). However, whether disrupted mCa2+ signaling contributes to neuronal pathology and cognitive decline independent of pre-existing amyloid or tau pathology remains unknown. Here, we generated mice with neuronal deletion of the mitochondrial sodium/calcium exchanger (NCLX, Slc8b1 gene), and evaluated age-associated changes in cognitive function and neuropathology. Neuronal loss of NCLX resulted in an age-dependent decline in spatial and cued recall memory, moderate amyloid deposition, mild tau pathology, synaptic remodeling, and indications of cell death. These results demonstrate that loss of NCLX-dependent mCa2+ efflux alone is sufficient to induce an Alzheimer’s disease-like pathology and highlights the promise of therapies targeting mCa2+ exchange.
- Published
- 2023
- Full Text
- View/download PDF
4. Editorial: Genomic alteration landscapes of aging, metabolic disorders, and cancer: Emerging overlaps and clinical importance
- Author
-
Jaspreet Kaur Dhanjal, Rajkumar Singh Kalra, Dhanendra Tomar, and Amrendra K. Ajay
- Subjects
genetic alterations ,cancer ,aging ,metabolic disorder ,mutations ,senescence ,Genetics ,QH426-470 - Published
- 2023
- Full Text
- View/download PDF
5. Yeast homologs of human MCUR1 regulate mitochondrial proline metabolism
- Author
-
Mohammad Zulkifli, John K. Neff, Shrishiv A. Timbalia, Natalie M. Garza, Yingqi Chen, Jeramie D. Watrous, Marta Murgia, Prachi P. Trivedi, Steven K. Anderson, Dhanendra Tomar, Roland Nilsson, Muniswamy Madesh, Mohit Jain, and Vishal M. Gohil
- Subjects
Science - Abstract
Although some fungal mitochondria lack the calcium uniporter, many intriguingly encode homologs of the uniporter assembly factor MCUR1. Here, the authors show that in budding yeast, the MCUR1 homologs Put6 and Put7 regulate mitochondrial proline metabolism, a function also conserved in human MCUR1.
- Published
- 2020
- Full Text
- View/download PDF
6. MICU1-dependent mitochondrial calcium uptake regulates lung alveolar type 2 cell plasticity and lung regeneration
- Author
-
Mir Ali, Xiaoying Zhang, Ryan LaCanna, Dhanendra Tomar, John W. Elrod, and Ying Tian
- Subjects
Cell biology ,Stem cells ,Medicine - Abstract
Lung alveolar type 2 (AT2) cells are progenitors for alveolar type 1 (AT1) cells. Although many factors regulate AT2 cell plasticity, the role of mitochondrial calcium (mCa2+) uptake in controlling AT2 cells remains unclear. We previously identified that the miR-302 family supports lung epithelial progenitor cell proliferation and less differentiated phenotypes during development. Here, we report that a sustained elevation of miR-302 in adult AT2 cells decreases AT2-to-AT1 cell differentiation during the Streptococcus pneumoniae–induced lung injury repair. We identified that miR-302 targets and represses the expression of mitochondrial Ca2+ uptake 1 (MICU1), which regulates mCa2+ uptake through the mCa2+ uniporter channel by acting as a gatekeeper at low cytosolic Ca2+ levels. Our results reveal a marked increase in MICU1 protein expression and decreased mCa2+ uptake during AT2-to-AT1 cell differentiation in the adult lung. Deletion of Micu1 in AT2 cells reduces AT2-to-AT1 cell differentiation during steady-state tissue maintenance and alveolar epithelial regeneration after bacterial pneumonia. These studies indicate that mCa2+ uptake is extensively modulated during AT2-to-AT1 cell differentiation and that MICU1-dependent mCa2+ uniporter channel gating is a prominent mechanism modulating AT2-to-AT1 cell differentiation.
- Published
- 2022
- Full Text
- View/download PDF
7. SARS-CoV-2 infection enhances mitochondrial PTP complex activity to perturb cardiac energetics
- Author
-
Karthik Ramachandran, Soumya Maity, Alagar R. Muthukumar, Soundarya Kandala, Dhanendra Tomar, Tarek Mohamed Abd El-Aziz, Cristel Allen, Yuyang Sun, Manigandan Venkatesan, Travis R. Madaris, Kevin Chiem, Rachel Truitt, Neelanjan Vishnu, Gregory Aune, Allen Anderson, Luis Martinez, Wenli Yang, James D. Stockand, Brij B. Singh, Subramanya Srikantan, W. Brian Reeves, and Muniswamy Madesh
- Subjects
Cardiovascular medicine ,Virology ,Transcriptomics ,Science - Abstract
Summary: SARS-CoV-2 is a newly identified coronavirus that causes the respiratory disease called coronavirus disease 2019 (COVID-19). With an urgent need for therapeutics, we lack a full understanding of the molecular basis of SARS-CoV-2-induced cellular damage and disease progression. Here, we conducted transcriptomic analysis of human PBMCs, identified significant changes in mitochondrial, ion channel, and protein quality-control gene products. SARS-CoV-2 proteins selectively target cellular organelle compartments, including the endoplasmic reticulum and mitochondria. M-protein, NSP6, ORF3A, ORF9C, and ORF10 bind to mitochondrial PTP complex components cyclophilin D, SPG-7, ANT, ATP synthase, and a previously undescribed CCDC58 (coiled-coil domain containing protein 58). Knockdown of CCDC58 or mPTP blocker cyclosporin A pretreatment enhances mitochondrial Ca2+ retention capacity and bioenergetics. SARS-CoV-2 infection exacerbates cardiomyocyte autophagy and promotes cell death that was suppressed by cyclosporin A treatment. Our findings reveal that SARS-CoV-2 viral proteins suppress cardiomyocyte mitochondrial function that disrupts cardiomyocyte Ca2+ cycling and cell viability.
- Published
- 2022
- Full Text
- View/download PDF
8. Mitochondrial calcium exchange links metabolism with the epigenome to control cellular differentiation
- Author
-
Alyssa A. Lombardi, Andrew A. Gibb, Ehtesham Arif, Devin W. Kolmetzky, Dhanendra Tomar, Timothy S. Luongo, Pooja Jadiya, Emma K. Murray, Pawel K. Lorkiewicz, György Hajnóczky, Elizabeth Murphy, Zoltan P. Arany, Daniel P. Kelly, Kenneth B. Margulies, Bradford G. Hill, and John W. Elrod
- Subjects
Science - Abstract
Myofibroblast differentiation contributes to extracellular matrix remodeling and fibrosis. Here, the authors report that alterations in mitochondrial calcium uptake is essential for metabolic reprogramming and epigenetic signaling for activation of the myofibroblast gene program.
- Published
- 2019
- Full Text
- View/download PDF
9. Impaired mitochondrial calcium efflux contributes to disease progression in models of Alzheimer’s disease
- Author
-
Pooja Jadiya, Devin W. Kolmetzky, Dhanendra Tomar, Antonio Di Meco, Alyssa A. Lombardi, Jonathan P. Lambert, Timothy S. Luongo, Marthe H. Ludtmann, Domenico Praticò, and John W. Elrod
- Subjects
Science - Abstract
Dysregulation of intracellular calcium is reported in Alzheimer’s disease. Here the authors show that loss of the mitochondrial Na+ /Ca2 + exchanger, NCLX – primary route of mitochondrial calcium efflux, precedes neuronal pathology in experimental models and contributes to Alzheimer’s disease progression.
- Published
- 2019
- Full Text
- View/download PDF
10. Mitochondrial dysfunction in human primary alveolar type II cells in emphysemaResearch in context
- Author
-
Beata Kosmider, Chih-Ru Lin, Loukmane Karim, Dhanendra Tomar, Liudmila Vlasenko, Nathaniel Marchetti, Sudhir Bolla, Muniswamy Madesh, Gerard J. Criner, and Karim Bahmed
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Cigarette smoke is the main risk factor of pulmonary emphysema development, which is characterized by alveolar wall destruction. Mitochondria are important for alveolar type II (ATII) cell metabolism due to ATP generation. Methods: We isolated ATII cells from control non-smoker and smoker organ donors, and after lung transplant of patients with emphysema to determine mitochondrial function, dynamics and mitochondrial (mt) DNA damage. Findings: We found high mitochondrial superoxide generation and mtDNA damage in ATII cells in emphysema. This correlated with decreased mtDNA amount. We also detected high TOP1-cc and low TDP1 levels in mitochondria in ATII cells in emphysema. This contributed to the decreased resolution of TOP1-cc leading to accumulation of mtDNA damage and mitochondrial dysfunction. Moreover, we used lung tissue obtained from areas with mild and severe emphysema from the same patients. We found a correlation between the impaired fusion and fission as indicated by low MFN1, OPA1, FIS1, and p-DRP1 levels and this disease severity. We detected lower TDP1 expression in severe compared to mild emphysema. Interpretation: We found high DNA damage and impairment of DNA damage repair in mitochondria in ATII cells isolated from emphysema patients, which contribute to abnormal mitochondrial dynamics. Our findings provide molecular mechanisms of mitochondrial dysfunction in this disease. Fund: This work was supported by National Institutes of Health (NIH) grant R01 HL118171 (B.K.) and the Catalyst Award from the American Lung Association (K.B.). Keywords: COPD, Emphysema, Lung, DNA damage, Alveolar type II cells, Mitochondria
- Published
- 2019
- Full Text
- View/download PDF
11. FOXD1-dependent MICU1 expression regulates mitochondrial activity and cell differentiation
- Author
-
Santhanam Shanmughapriya, Dhanendra Tomar, Zhiwei Dong, Katherine J. Slovik, Neeharika Nemani, Kalimuthusamy Natarajaseenivasan, Edmund Carvalho, Christy Lu, Kaitlyn Corrigan, Venkata Naga Srikanth Garikipati, Jessica Ibetti, Sudarsan Rajan, Carlos Barrero, Kurt Chuprun, Raj Kishore, Salim Merali, Ying Tian, Wenli Yang, and Muniswamy Madesh
- Subjects
Science - Abstract
Genetic ablation of Mitochondrial Ca2+ uptake protein 1 (MICU1) in mouse induces higher rates of perinatal lethality. Here the authors show that MICU1 expression is regulated by hypoxia in a FOXD1-dependent manner, establishing a cyclic switch between glycolytic and oxidative metabolism during development.
- Published
- 2018
- Full Text
- View/download PDF
12. MIRO-1 Determines Mitochondrial Shape Transition upon GPCR Activation and Ca2+ Stress
- Author
-
Neeharika Nemani, Edmund Carvalho, Dhanendra Tomar, Zhiwei Dong, Andrea Ketschek, Sarah L. Breves, Fabián Jaña, Alison M. Worth, Julie Heffler, Palaniappan Palaniappan, Aparna Tripathi, Ramasamy Subbiah, Massimo F. Riitano, Ajay Seelam, Thomas Manfred, Kie Itoh, Shuxia Meng, Hiromi Sesaki, William J. Craigen, Sudarsan Rajan, Santhanam Shanmughapriya, Jeffrey Caplan, Benjamin L. Prosser, Donald L. Gill, Peter B. Stathopulos, Gianluca Gallo, David C. Chan, Prashant Mishra, and Muniswamy Madesh
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Mitochondria shape cytosolic calcium ([Ca2+]c) transients and utilize the mitochondrial Ca2+ ([Ca2+]m) in exchange for bioenergetics output. Conversely, dysregulated [Ca2+]c causes [Ca2+]m overload and induces permeability transition pore and cell death. Ablation of MCU-mediated Ca2+ uptake exhibited elevated [Ca2+]c and failed to prevent stress-induced cell death. The mechanisms for these effects remain elusive. Here, we report that mitochondria undergo a cytosolic Ca2+-induced shape change that is distinct from mitochondrial fission and swelling. [Ca2+]c elevation, but not MCU-mediated Ca2+ uptake, appears to be essential for the process we term mitochondrial shape transition (MiST). MiST is mediated by the mitochondrial protein Miro1 through its EF-hand domain 1 in multiple cell types. Moreover, Ca2+-dependent disruption of Miro1/KIF5B/tubulin complex is determined by Miro1 EF1 domain. Functionally, Miro1-dependent MiST is essential for autophagy/mitophagy that is attenuated in Miro1 EF1 mutants. Thus, Miro1 is a cytosolic Ca2+ sensor that decodes metazoan Ca2+ signals as MiST. : Metazoan Ca2+ signal determines mitochondrial shape transition (MiST) and cellular quality control. Nemani et al. find that mitochondria undergo shape changes upon Ca2+ stress. MiST is distinct from matrix Ca2+-induced swelling and mitochondrial dynamics. The conserved Ca2+ sensor Miro1 enables MiST and promotes autophagy/mitophagy. Keywords: mitochondrial shape, MiST, calcium, Miro, EF hand, PTP, MCU, mitophagy, autophagy, mitochondrial dynamics
- Published
- 2018
- Full Text
- View/download PDF
13. Blockade of MCU-Mediated Ca2+ Uptake Perturbs Lipid Metabolism via PP4-Dependent AMPK Dephosphorylation
- Author
-
Dhanendra Tomar, Fabián Jaña, Zhiwei Dong, William J. Quinn, III, Pooja Jadiya, Sarah L. Breves, Cassidy C. Daw, Subramanya Srikantan, Santhanam Shanmughapriya, Neeharika Nemani, Edmund Carvalho, Aparna Tripathi, Alison M. Worth, Xueqian Zhang, Roshanak Razmpour, Ajay Seelam, Stephen Rhode, Anuj V. Mehta, Michael Murray, Daniel Slade, Servio H. Ramirez, Prashant Mishra, Glenn S. Gerhard, Jeffrey Caplan, Luke Norton, Kumar Sharma, Sudarsan Rajan, Darius Balciunas, Dayanjan S. Wijesinghe, Rexford S. Ahima, Joseph A. Baur, and Muniswamy Madesh
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Mitochondrial Ca2+ uniporter (MCU)-mediated Ca2+ uptake promotes the buildup of reducing equivalents that fuel oxidative phosphorylation for cellular metabolism. Although MCU modulates mitochondrial bioenergetics, its function in energy homeostasis in vivo remains elusive. Here we demonstrate that deletion of the Mcu gene in mouse liver (MCUΔhep) and in Danio rerio by CRISPR/Cas9 inhibits mitochondrial Ca2+ (mCa2+) uptake, delays cytosolic Ca2+ (cCa2+) clearance, reduces oxidative phosphorylation, and leads to increased lipid accumulation. Elevated hepatic lipids in MCUΔhep were a direct result of extramitochondrial Ca2+-dependent protein phosphatase-4 (PP4) activity, which dephosphorylates AMPK. Loss of AMPK recapitulates hepatic lipid accumulation without changes in MCU-mediated Ca2+ uptake. Furthermore, reconstitution of active AMPK, or PP4 knockdown, enhances lipid clearance in MCUΔhep hepatocytes. Conversely, gain-of-function MCU promotes rapid mCa2+ uptake, decreases PP4 levels, and reduces hepatic lipid accumulation. Thus, our work uncovers an MCU/PP4/AMPK molecular cascade that links Ca2+ dynamics to hepatic lipid metabolism. : Hepatic mitochondrial Ca2+ shapes bioenergetics and lipid homeostasis. Tomar et al. demonstrate that MCU-mediated cCa2+ buffering serves as a crucial step in controlling hepatic fuel metabolism through an MCU/PP4/AMPK molecular cascade. Identification of these molecular signaling events aids in understanding how perturbation of mitochondrial ion homeostasis may contribute to the etiology of metabolic disorders. Keywords: mitochondrial Ca2+ uniporter, calcium, bioenergetics, AMPK, MCU, hepatocyte, lipid metabolism, phosphatase, metabolic diseases, diabetes
- Published
- 2019
- Full Text
- View/download PDF
14. MCUR1 Is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial Bioenergetics
- Author
-
Dhanendra Tomar, Zhiwei Dong, Santhanam Shanmughapriya, Diana A. Koch, Toby Thomas, Nicholas E. Hoffman, Shrishiv A. Timbalia, Samuel J. Goldman, Sarah L. Breves, Daniel P. Corbally, Neeharika Nemani, Joseph P. Fairweather, Allison R. Cutri, Xueqian Zhang, Jianliang Song, Fabián Jaña, Jianhe Huang, Carlos Barrero, Joseph E. Rabinowitz, Timothy S. Luongo, Sarah M. Schumacher, Michael E. Rockman, Alexander Dietrich, Salim Merali, Jeffrey Caplan, Peter Stathopulos, Rexford S. Ahima, Joseph Y. Cheung, Steven R. Houser, Walter J. Koch, Vickas Patel, Vishal M. Gohil, John W. Elrod, Sudarsan Rajan, and Muniswamy Madesh
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Mitochondrial Ca2+ Uniporter (MCU)-dependent mitochondrial Ca2+ uptake is the primary mechanism for increasing matrix Ca2+ in most cell types. However, a limited understanding of the MCU complex assembly impedes the comprehension of the precise mechanisms underlying MCU activity. Here, we report that mouse cardiomyocytes and endothelial cells lacking MCU regulator 1 (MCUR1) have severely impaired [Ca2+]m uptake and IMCU current. MCUR1 binds to MCU and EMRE and function as a scaffold factor. Our protein binding analyses identified the minimal, highly conserved regions of coiled-coil domain of both MCU and MCUR1 that are necessary for heterooligomeric complex formation. Loss of MCUR1 perturbed MCU heterooligomeric complex and functions as a scaffold factor for the assembly of MCU complex. Vascular endothelial deletion of MCU and MCUR1 impaired mitochondrial bioenergetics, cell proliferation, and migration but elicited autophagy. These studies establish the existence of a MCU complex that assembles at the mitochondrial integral membrane and regulates Ca2+-dependent mitochondrial metabolism.
- Published
- 2016
- Full Text
- View/download PDF
15. SARS-CoV-2, ACE2, and Hydroxychloroquine: Cardiovascular Complications, Therapeutics, and Clinical Readouts in the Current Settings
- Author
-
Rajkumar Singh Kalra, Dhanendra Tomar, Avtar Singh Meena, and Ramesh Kandimalla
- Subjects
SARS-CoV-2 ,COVID-19 ,ACE2 ,hydroxychloroquine ,cardiovascular system ,cardiovascular disease (CVD) ,Medicine - Abstract
The rapidly evolving coronavirus disease 2019 (COVID-19, caused by severe acute respiratory syndrome coronavirus 2- SARS-CoV-2), has greatly burdened the global healthcare system and led it into crisis in several countries. Lack of targeted therapeutics led to the idea of repurposing broad-spectrum drugs for viral intervention. In vitro analyses of hydroxychloroquine (HCQ)’s anecdotal benefits prompted its widespread clinical repurposing globally. Reports of emerging cardiovascular complications due to its clinical prescription are revealing the crucial role of angiotensin-converting enzyme 2 (ACE2), which serves as a target receptor for SARS-CoV-2. In the present settings, a clear understanding of these targets, their functional aspects and physiological impact on cardiovascular function are critical. In an up-to-date format, we shed light on HCQ’s anecdotal function in stalling SARS-CoV-2 replication and immunomodulatory activities. While starting with the crucial role of ACE2, we here discuss the impact of HCQ on systemic cardiovascular function, its associated risks, and the scope of HCQ-based regimes in current clinical settings. Citing the extent of HCQ efficacy, the key considerations and recommendations for the use of HCQ in clinics are further discussed. Taken together, this review provides crucial insights into the role of ACE2 in SARS-CoV-2-led cardiovascular activity, and concurrently assesses the efficacy of HCQ in contemporary clinical settings.
- Published
- 2020
- Full Text
- View/download PDF
16. Nucleo-cytoplasmic trafficking of TRIM8, a novel oncogene, is involved in positive regulation of TNF induced NF-κB pathway.
- Author
-
Dhanendra Tomar, Lakshmi Sripada, Paresh Prajapati, Rochika Singh, Arun Kumar Singh, and Rajesh Singh
- Subjects
Medicine ,Science - Abstract
TNF induced nuclear factor kappa B (NF-κB) is one of the central signaling pathways that plays a critical role in carcinogenesis and inflammatory diseases. Post-translational modification through ubiquitin plays important role in the regulation of this pathway. In the current study, we investigated the role of TRIM8, member of RING family ubiquitin ligase in regulation of NF-κB pathway. We observed that TRIM8 positively regulates TNF induced NF-κB pathway. Different domains of TRIM8 showed discrete functions at the different steps in regulation of TNF induced NF-κB pathway. Ubiquitin ligase activity of TRIM8 is essential for regulation of NF-κB activation in both cytoplasm as well as nucleus. TRIM8 negates PIAS3 mediated negative repression of NF-κB at p65 by inducing translocation of PIAS3 from nucleus to cytoplasm as well as its turnover. TNF induces translocation of TRIM8 from nucleus to cytoplasm, which positively regulates NF-κB. The cytoplasmic translocation of TRIM8 is essential for TNF induced NF-κB but not for p65 mediated NF-κB regulation. TRIM8 also enhanced the clonogenic and migration ability of cells by modulating NF-κB. The further study will help to understand the role of TRIM8 in inflammation and cancer.
- Published
- 2012
- Full Text
- View/download PDF
17. Systematic analysis of small RNAs associated with human mitochondria by deep sequencing: detailed analysis of mitochondrial associated miRNA.
- Author
-
Lakshmi Sripada, Dhanendra Tomar, Paresh Prajapati, Rochika Singh, Arun Kumar Singh, and Rajesh Singh
- Subjects
Medicine ,Science - Abstract
Mitochondria are one of the central regulators of many cellular processes beyond its well established role in energy metabolism. The inter-organellar crosstalk is critical for the optimal function of mitochondria. Many nuclear encoded proteins and RNA are imported to mitochondria. The translocation of small RNA (sRNA) including miRNA to mitochondria and other sub-cellular organelle is still not clear. We characterized here sRNA including miRNA associated with human mitochondria by cellular fractionation and deep sequencing approach. Mitochondria were purified from HEK293 and HeLa cells for RNA isolation. The sRNA library was generated and sequenced using Illumina system. The analysis showed the presence of unique population of sRNA associated with mitochondria including miRNA. Putative novel miRNAs were characterized from unannotated sRNA sequences. The study showed the association of 428 known, 196 putative novel miRNAs to mitochondria of HEK293 and 327 known, 13 putative novel miRNAs to mitochondria of HeLa cells. The alignment of sRNA to mitochondrial genome was also studied. The targets were analyzed using DAVID to classify them in unique networks using GO and KEGG tools. Analysis of identified targets showed that miRNA associated with mitochondria regulates critical cellular processes like RNA turnover, apoptosis, cell cycle and nucleotide metabolism. The six miRNAs (counts >1000) associated with mitochondria of both HEK293 and HeLa were validated by RT-qPCR. To our knowledge, this is the first systematic study demonstrating the associations of sRNA including miRNA with mitochondria that may regulate site-specific turnover of target mRNA important for mitochondrial related functions.
- Published
- 2012
- Full Text
- View/download PDF
18. Nicotinamide riboside kinase-2 regulates metabolic adaptation in the ischemic heart
- Author
-
Hezlin Marzook, Anamika Gupta, Dhanendra Tomar, Mohamed A. Saleh, Kiran Patil, Mohammad H. Semreen, Rifat Hamoudi, Nelson C. Soares, Rizwan Qaisar, and Firdos Ahmad
- Subjects
Drug Discovery ,Molecular Medicine ,Genetics (clinical) - Published
- 2023
- Full Text
- View/download PDF
19. MICU1 regulates mitochondrial cristae structure and function independently of the mitochondrial Ca 2+ uniporter channel
- Author
-
Dhanendra Tomar, Manfred Thomas, Joanne F. Garbincius, Devin W. Kolmetzky, Oniel Salik, Pooja Jadiya, Suresh K. Joseph, April C. Carpenter, György Hajnóczky, and John W. Elrod
- Subjects
Cell Biology ,Molecular Biology ,Biochemistry - Abstract
MICU1 is a calcium (Ca 2+ )–binding protein that regulates the mitochondrial Ca 2+ uniporter channel complex (mtCU) and mitochondrial Ca 2+ uptake. MICU1 knockout mice display disorganized mitochondrial architecture, a phenotype that is distinct from that of mice with deficiencies in other mtCU subunits and, thus, is likely not explained by changes in mitochondrial matrix Ca 2+ content. Using proteomic and cellular imaging techniques, we found that MICU1 localized to the mitochondrial contact site and cristae organizing system (MICOS) and directly interacted with the MICOS components MIC60 and CHCHD2 independently of the mtCU. We demonstrated that MICU1 was essential for MICOS complex formation and that MICU1 ablation resulted in altered cristae organization, mitochondrial ultrastructure, mitochondrial membrane dynamics, and cell death signaling. Together, our results suggest that MICU1 is an intermembrane space Ca 2+ sensor that modulates mitochondrial membrane dynamics independently of matrix Ca 2+ uptake. This system enables distinct Ca 2+ signaling in the mitochondrial matrix and at the intermembrane space to modulate cellular energetics and cell death in a concerted manner.
- Published
- 2023
- Full Text
- View/download PDF
20. Genomic alteration landscapes of aging, metabolic disorders, and cancer: emerging overlaps and clinical importance
- Author
-
Rajkumar S. Kalra, Amrendra K. Ajay, Dhanendra Tomar, and Jaspreet Kaur Dhanjal
- Published
- 2023
- Full Text
- View/download PDF
21. Molecular Signature of HFpEF
- Author
-
John W. Elrod, Steven R. Houser, Markus Wallner, Remus M. Berretta, Dhanendra Tomar, Anh T Huynh, Joanne F Garbincius, Deborah M Eaton, Emma K Murray, Devin W. Kolmetzky, and Andrew A. Gibb
- Subjects
MS/MS, tandem mass spectrometry ,FDR, false discovery rate ,Systems biology ,ETC, electron transport chain ,heart failure ,HFpEF, heart failure with preserved ejection fraction ,Mitochondrion ,Biology ,HF, heart failure ,UPLC, ultraperformance liquid chromatography ,Transcriptome ,transcriptomics ,Metabolomics ,medicine ,EF, ejection fraction ,LA, left atrial ,LAV, left atrial volume ,m/z, mass to charge ratio ,HFrEF, heart failure with reduced ejection fraction ,GO, gene ontology ,Maladaptation ,ESI, electrospray ionization ,LV, left ventricle/ventricular ,Skeletal muscle ,systems biology ,FC, fold change ,medicine.disease ,metabolomics ,preserved ejection fraction ,ECM, extracellular matrix ,Cell biology ,mitochondria ,BCAA, branched chain amino acids ,medicine.anatomical_structure ,RI, retention index ,Heart failure ,Preclinical Research ,RCR, respiratory control ratio ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,DAG, diacylglycerol - Abstract
Visual Abstract, Highlights • Early cardiac mitochondrial dysfunction is mediated by transcriptional down-regulation of the mitochondrial proteome. • Comprehensive metabolic remodeling is conserved throughout HFpEF progression and includes increased amino acid and lipid species, indicative of impaired oxidative metabolism. • Transcriptional and metabolic remodeling of skeletal muscle suggests cardiac signaling as a mediator of peripheral tissue maladaptation. • Unbiased systems-level analysis provides new mechanisms underlying HFpEF development., Summary In this study the authors used systems biology to define progressive changes in metabolism and transcription in a large animal model of heart failure with preserved ejection fraction (HFpEF). Transcriptomic analysis of cardiac tissue, 1-month post-banding, revealed loss of electron transport chain components, and this was supported by changes in metabolism and mitochondrial function, altogether signifying alterations in oxidative metabolism. Established HFpEF, 4 months post-banding, resulted in changes in intermediary metabolism with normalized mitochondrial function. Mitochondrial dysfunction and energetic deficiencies were noted in skeletal muscle at early and late phases of disease, suggesting cardiac-derived signaling contributes to peripheral tissue maladaptation in HFpEF. Collectively, these results provide insights into the cellular biology underlying HFpEF progression.
- Published
- 2021
- Full Text
- View/download PDF
22. Suppression of Ca 2+ signaling enhances melanoma progression
- Author
-
Scott Gross, Robert Hooper, Dhanendra Tomar, Alexander P Armstead, No'ad Shanas, Pranava Mallu, Hinal Joshi, Suravi Ray, Parkson Lee‐Gau Chong, Igor Astsaturov, Jeffrey M Farma, Kathy Q Cai, Kumaraswamy Naidu Chitrala, John W Elrod, M Raza Zaidi, and Jonathan Soboloff
- Subjects
General Immunology and Microbiology ,General Neuroscience ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
23. Abstract P1036: Role Of Mitochondrial Ribosomal Protein L7/l12 (mrpl12) In Diabetic Ischemic Heart Disease
- Author
-
Amit K Rai, Brooke Lee, Shridhar Sanghvi, Dhanendra Tomar, Dhananjie Chandrasekera, Sreejit Gopala Krishna, Devasena Ponnalagu, Mahmood Khan, Harpreet Singh, Prabhakara R Nagareddy, David A Goukassian, Rajesh Katare, Walter J Koch, Raj Kishore, and Venkata Garikipati
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Myocardial infarction (MI) is a significant cause of death in diabetic patients. In addition, growing evidence suggests that mitochondrial dysfunction contributes to heart failure in diabetes. However, the molecular mechanisms of mitochondrial dysfunction mediating heart failure in diabetes are still poorly understood. The current study aimed to investigate the role of mitochondrial ribosomal protein L7/L12 (MRPL12) in mouse models of type II diabetes (db/db mice)and high-fat diet (HFD) mice with or without induction of MI and human hearts with or without diabetes (n=7) .Data analysis revealed an increase in MRPL12 levels in the LV tissue of HFD fed mice with MI than in LV tissues of low-fat diet-fed mice with MI, whereas MRPL12 levels remained unchanged in the db/db mice with MI. Intriguingly, we found increased MRPL12 levels in atrial appendage tissue of diabetic patients with ischemic heart disease compared to non-diabetic patients. We utilized human cardiomyocyte cell-line (AC-16) as surrogate models to delineate the mechanisms; surprisingly, adenovirus-mediated overexpression of MRPL12 with or without hyperglycemia in AC-16 cardiomyocytes does not affect mitochondrial OXPHOS . In addition, overexpression of MRPL12 had no effect on the mitochondrial ROS, mitochondrial membrane depolarization, and caspase activity in AC-16 cardiomyocytes. Whereas RNA interference (RNAi)-mediated MRPL12 silencing remarkedly reduced mitochondrial oxidative phosphorylation in AC-16 cells without any stress. In addition, knockdown of MRPL12 increased mitochondrial membrane depolarization mitochondrial ROS and reduced maximal respiratory capacity of mitochondria without any stress. Overall, our results provide new insights into the role of MRPL12 in the pathophysiology of MI in diabetes.
- Published
- 2022
- Full Text
- View/download PDF
24. Suppression of Ca
- Author
-
Scott, Gross, Robert, Hooper, Dhanendra, Tomar, Alexander P, Armstead, No'ad, Shanas, Pranava, Mallu, Hinal, Joshi, Suravi, Ray, Parkson Lee-Gau, Chong, Igor, Astsaturov, Jeffrey M, Farma, Kathy Q, Cai, Kumaraswamy Naidu, Chitrala, John W, Elrod, M Raza, Zaidi, and Jonathan, Soboloff
- Subjects
Cholesterol ,Glucose ,ORAI1 Protein ,Humans ,Calcium ,Calcium Channels ,Calcium Signaling ,Stromal Interaction Molecule 1 ,Melanoma - Abstract
The role of store-operated Ca
- Published
- 2022
25. TMBIM5 loss of function alters mitochondrial matrix ion homeostasis and causes a skeletal myopathy
- Author
-
Li Zhang, Felicia Dietsche, Bruno Seitaj, Liliana Rojas-Charry, Nadina Latchman, Dhanendra Tomar, Rob CI Wüst, Alexander Nickel, Katrin BM Frauenknecht, Benedikt Schoser, Sven Schumann, Michael J Schmeisser, Johannes vom Berg, Thorsten Buch, Stefanie Finger, Philip Wenzel, Christoph Maack, John W Elrod, Jan B Parys, Geert Bultynck, Axel Methner, University of Zurich, Methner, Axel, Physiology, AMS - Ageing & Vitality, and AMS - Musculoskeletal Health
- Subjects
Ecology ,Health, Toxicology and Mutagenesis ,610 Medizin ,610 Medicine & health ,Apoptosis ,Plant Science ,Genetics and Molecular Biology (miscellaneous) ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,1301 Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,Mitochondrial Membrane Transport Proteins ,Mice ,Muscular Diseases ,Health ,610 Medical sciences ,1110 Plant Science ,2307 Health, Toxicology and Mutagenesis ,570 Life sciences ,biology ,590 Animals (Zoology) ,Animals ,Homeostasis ,10239 Institute of Laboratory Animal Science ,Toxicology and Mutagenesis ,Calcium ,2303 Ecology - Abstract
Ion fluxes across the inner mitochondrial membrane control mitochondrial volume, energy production, and apoptosis. TMBIM5, a highly conserved protein with homology to putative pH-dependent ion channels, is involved in the maintenance of mitochondrial cristae architecture, ATP production, and apoptosis. Here, we demonstrate that overexpressed TMBIM5 can mediate mitochondrial calcium uptake. Under steady-state conditions, loss of TMBIM5 results in increased potassium and reduced proton levels in the mitochondrial matrix caused by attenuated exchange of these ions. To identify the in vivo consequences of TMBIM5 dysfunction, we generated mice carrying a mutation in the channel pore. These mutant mice display increased embryonic or perinatal lethality and a skeletal myopathy which strongly correlates with tissue-specific disruption of cristae architecture, early opening of the mitochondrial permeability transition pore, reduced calcium uptake capability, and mitochondrial swelling. Our results demonstrate that TMBIM5 is an essential and important part of the mitochondrial ion transport system machinery with particular importance for embryonic development and muscle function. ispartof: LIFE SCIENCE ALLIANCE vol:5 issue:10 ispartof: location:United States status: published
- Published
- 2022
- Full Text
- View/download PDF
26. Novel BAG3 Variants in African American Patients With Cardiomyopathy: Reduced β-Adrenergic Responsiveness in Excitation–Contraction
- Author
-
Joseph Y. Cheung, Jianliang Song, Kamel Khalili, Arthur M. Feldman, Jennifer Gordon, Xue-Qian Zhang, Dhanendra Tomar, JuFang Wang, Glenn S. Gerhard, and Valerie D. Myers
- Subjects
BAG domain ,medicine.medical_specialty ,Contraction (grammar) ,medicine.medical_treatment ,Cardiomyopathy ,030204 cardiovascular system & hematology ,BAG3 ,Article ,WW domain ,Mice ,03 medical and health sciences ,Adrenergic Agents ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Myocyte ,Myocytes, Cardiac ,030212 general & internal medicine ,Adaptor Proteins, Signal Transducing ,Heart Failure ,Heart transplantation ,biology ,business.industry ,Isoproterenol ,medicine.disease ,Myocardial Contraction ,Black or African American ,Endocrinology ,Heart failure ,biology.protein ,Apoptosis Regulatory Proteins ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background We reported 3 novel nonsynonymous single nucleotide variants of Bcl2-associated athanogene 3 (BAG3) in African Americans with heart failure (HF) that are associated with a 2-fold increase in cardiac events (HF hospitalization, heart transplantation, or death). Methods and Results We expressed BAG3 variants (P63A, P380S, and A479V) via adenovirus-mediated gene transfer in adult left ventricular myocytes isolated from either wild-type (WT) or cardiac-specific BAG3 haploinsufficient (cBAG3+/−) mice: the latter to simulate the clinical situation in which BAG3 variants are only found on 1 allele. Compared with WT myocytes, cBAG3+/− myocytes expressed approximately 50% of endogenous BAG3 levels and exhibited decreased [Ca2+]i and contraction amplitudes after isoproterenol owing to decreased L-type Ca2+ current. BAG3 repletion with WT BAG3 but not P380S, A479V, or P63A/P380S variants restored contraction amplitudes in cBAG3+/− myocytes to those measured in WT myocytes, suggesting excitation–contraction abnormalities partly account for HF in patients harboring these mutants. Because P63A is near the WW domain (residues 21–55) and A479V is in the BAG domain (residues 420–499), we expressed BAG3 deletion mutants (Δ1–61 and Δ421–575) in WT myocytes and demonstrated that the BAG but not the WW domain was involved in enhancement of excitation–contraction by isoproterenol. Conclusions The BAG3 variants contribute to HF in African American patients partly by decreasing myocyte excitation–contraction under stress, and that both the BAG and PXXP domains are involved in mediating β-adrenergic responsiveness in myocytes.
- Published
- 2020
- Full Text
- View/download PDF
27. The role of DJ-1 in human primary alveolar type II cell injury induced by e-cigarette aerosol
- Author
-
Ellen M. Unterwald, Dhanendra Tomar, Elise M. Messier, Muniswamy Madesh, Mark O. Aksoy, Robert J. Mason, Gerard J. Criner, John W. Elrod, Toby K. Eisenstein, Karim Bahmed, Loukmane Karim, Hannah Simborio, Chih-Ru Lin, Steven G. Kelsen, and Beata Kosmider
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Nicotine ,Physiology ,Primary Cell Culture ,Protein Deglycase DJ-1 ,Apoptosis ,Electronic Nicotine Delivery Systems ,Mitochondrion ,Oxidative Phosphorylation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Superoxides ,Physiology (medical) ,medicine ,Animals ,Humans ,Alveolar type II cell ,Aerosols ,Membrane Potential, Mitochondrial ,Mice, Knockout ,Lung ,Primary (chemistry) ,Chemistry ,Interleukin-8 ,Cell Biology ,respiratory system ,Molecular biology ,Mitochondria ,Aerosol ,Pulmonary Alveoli ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Alveolar Epithelial Cells ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Calcium ,DNA Damage ,Research Article - Abstract
The alveolus participates in gas exchange, which can be impaired by environmental factors and toxins. There is an increase in using electronic cigarettes (e-cigarettes); however, their effect on human primary alveolar epithelial cells is unknown. Human lungs were obtained from nonsmoker organ donors to isolate alveolar type II (ATII) cells. ATII cells produce and secrete pulmonary surfactant and restore the epithelium after damage, and mitochondrial function is important for their metabolism. Our data indicate that human ATII cell exposure to e-cigarette aerosol increased IL-8 levels and induced DNA damage and apoptosis. We also studied the cytoprotective effect of DJ-1 against ATII cell injury. DJ-1 knockdown in human primary ATII cells sensitized cells to mitochondrial dysfunction as detected by high mitochondrial superoxide production, decreased mitochondrial membrane potential, and calcium elevation. DJ-1 knockout (KO) mice were more susceptible to ATII cell apoptosis and lung injury induced by e-cigarette aerosol compared with wild-type mice. Regulation of the oxidative phosphorylation (OXPHOS) is important for mitochondrial function and protection against oxidative stress. Major subunits of the OXPHOS system are encoded by both nuclear and mitochondrial DNA. We found dysregulation of OXPHOS complexes in DJ-1 KO mice after exposure to e-cigarette aerosol, which could disrupt the nuclear/mitochondrial stoichiometry, resulting in mitochondrial dysfunction. Together, our results indicate that DJ-1 deficiency sensitizes ATII cells to damage induced by e-cigarette aerosol leading to lung injury.
- Published
- 2019
- Full Text
- View/download PDF
28. Mitochondrial dysfunction in human primary alveolar type II cells in emphysema
- Author
-
Sudhir Bolla, Dhanendra Tomar, Karim Bahmed, Loukmane Karim, Beata Kosmider, Muniswamy Madesh, Liudmila Vlasenko, Gerard J. Criner, Chih-Ru Lin, and Nathaniel Marchetti
- Subjects
0301 basic medicine ,FIS1 ,Mitochondrial DNA ,Pathology ,medicine.medical_specialty ,Research paper ,DNA damage ,Mitochondrion ,DNA, Mitochondrial ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,Superoxides ,Smoke ,Humans ,COPD ,Medicine ,MFN1 ,Lung ,Emphysema ,Phosphoric Diester Hydrolases ,business.industry ,General Medicine ,respiratory system ,medicine.disease ,Mitochondria ,Alveolar type II cells ,respiratory tract diseases ,3. Good health ,Oxidative Stress ,Protein Transport ,030104 developmental biology ,medicine.anatomical_structure ,Pulmonary Emphysema ,Alveolar Epithelial Cells ,030220 oncology & carcinogenesis ,Disease Progression ,Energy Metabolism ,Reactive Oxygen Species ,business ,TDP1 - Abstract
Background Cigarette smoke is the main risk factor of pulmonary emphysema development, which is characterized by alveolar wall destruction. Mitochondria are important for alveolar type II (ATII) cell metabolism due to ATP generation. Methods We isolated ATII cells from control non-smoker and smoker organ donors, and after lung transplant of patients with emphysema to determine mitochondrial function, dynamics and mitochondrial (mt) DNA damage. Findings We found high mitochondrial superoxide generation and mtDNA damage in ATII cells in emphysema. This correlated with decreased mtDNA amount. We also detected high TOP1-cc and low TDP1 levels in mitochondria in ATII cells in emphysema. This contributed to the decreased resolution of TOP1-cc leading to accumulation of mtDNA damage and mitochondrial dysfunction. Moreover, we used lung tissue obtained from areas with mild and severe emphysema from the same patients. We found a correlation between the impaired fusion and fission as indicated by low MFN1, OPA1, FIS1, and p-DRP1 levels and this disease severity. We detected lower TDP1 expression in severe compared to mild emphysema. Interpretation We found high DNA damage and impairment of DNA damage repair in mitochondria in ATII cells isolated from emphysema patients, which contribute to abnormal mitochondrial dynamics. Our findings provide molecular mechanisms of mitochondrial dysfunction in this disease. Fund This work was supported by National Institutes of Health (NIH) grant R01 HL118171 (B.K.) and the Catalyst Award from the American Lung Association (K.B.).
- Published
- 2019
- Full Text
- View/download PDF
29. Trpm2 enhances physiological bioenergetics and protects against pathological oxidative cardiac injury: Role of Pyk2 phosphorylation
- Author
-
Muniswamy Madesh, Sudasan Rajan, Santhanam Shanmughapriya, Shey-Shing Sheu, Barbara A. Miller, Xue-Qian Zhang, Iwona Hirschler-Laszkiewicz, Arthur M. Feldman, Jianliang Song, JuFang Wang, Dhanendra Tomar, and Joseph Y. Cheung
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Reactive oxygen species ,Physiology ,Chemistry ,Superoxide ,Clinical Biochemistry ,Cell Biology ,Oxidative phosphorylation ,Mitochondrion ,Article ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,BAPTA ,030220 oncology & carcinogenesis ,Ca2+/calmodulin-dependent protein kinase ,Phosphorylation ,Adenosine triphosphate - Abstract
The mechanisms by which Trpm2 channels enhance mitochondrial bioenergetics and protect against oxidative stress-induced cardiac injury remain unclear. Here, the role of proline-rich tyrosine kinase 2 (Pyk2) in Trpm2 signaling is explored. Activation of Trpm2 in adult myocytes with H2 O2 resulted in 10- to 21-fold increases in Pyk2 phosphorylation in wild-type (WT) myocytes which was significantly lower (~40%) in Trpm2 knockout (KO) myocytes. Pyk2 phosphorylation was inhibited (~54%) by the Trpm2 blocker clotrimazole. Buffering Trpm2-mediated Ca2+ increase with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) resulted in significantly reduced pPyk2 in WT but not in KO myocytes, indicating Ca2+ influx through activated Trpm2 channels phosphorylated Pyk2. Part of phosphorylated Pyk2 translocated from cytosol to mitochondria which has been previously shown to augment mitochondrial Ca2+ uptake and enhance adenosine triphosphate generation. Although Trpm2-mediated Ca2+ influx phosphorylated Ca2+ -calmodulin kinase II (CaMKII), the CaMKII inhibitor KN93 did not significantly affect Pyk2 phosphorylation in H2 O2 -treated WT myocytes. After ischemia/reperfusion (I/R), Pyk2 phosphorylation and its downstream prosurvival signaling molecules (pERK1/2 and pAkt) were significantly lower in KO-I/R when compared with WT-I/R hearts. After hypoxia/reoxygenation, mitochondrial membrane potential was lower and superoxide level was higher in KO myocytes, and were restored to WT values by the mitochondria-targeted superoxide scavenger MitoTempo. Our results suggested that Ca2+ influx via tonically activated Trpm2 phosphorylated Pyk2, part of which translocated to mitochondria, resulting in better mitochondrial bioenergetics to maintain cardiac health. After I/R, Pyk2 activated prosurvival signaling molecules and prevented excessive increases in reactive oxygen species, thereby affording protection from I/R injury.
- Published
- 2019
- Full Text
- View/download PDF
30. Neuronal Loss of NCLX-Dependent Mitochondrial Calcium Efflux Contributes to Age-Associated Cognitive Decline
- Author
-
Devin W. Kolmetzky, Dhanendra Tomar, John W. Elrod, Henry M. Cohen, and Pooja Jadiya
- Subjects
History ,Polymers and Plastics ,Mechanism (biology) ,Amyloidosis ,chemistry.chemical_element ,Cognition ,Neuropathology ,Disease ,Calcium ,Biology ,medicine.disease ,Industrial and Manufacturing Engineering ,chemistry ,medicine ,Efflux ,Business and International Management ,Cognitive decline ,Neuroscience - Abstract
Mitochondrial and metabolic impairments contribute to neurodegenerative disease development. We recently reported that mitochondrial calcium (mCa2+) overload due to remodeling of the mitochondrial calcium exchange machinery is a causal determinant of cognitive decline in Alzheimer’s disease models. However, whether disrupted mCa2+ signaling contributes to neuronal pathology and cognitive decline independent of precedent amyloidosis or tau pathology remains unknown. Here, we generated mice with neuronal-specific deletion of the mitochondrial sodium/calcium exchanger (NCLX), the primary mechanism of mCa2+ efflux, and evaluated age-associated changes in cognitive function and neuropathology. Neuronal- loss of NCLX resulted in an age-dependent decline in spatial and cued recall memory, moderate amyloid deposition, mild tau hyperphosphorylation, and synaptic remodeling. These results demonstrate that loss of NCLX-dependent mCa2+ efflux alone is sufficient to induce an Alzheimer’s disease-like pathology and highlights the promise of therapies targeting mCa2+ exchange.
- Published
- 2021
- Full Text
- View/download PDF
31. Genetic Ablation of Neuronal Mitochondrial Calcium Uptake Halts Alzheimer's Disease Progression
- Author
-
Manfred Thomas, Dhanendra Tomar, Pooja Jadiya, Salman Khaledi, Joanne F Garbincius, Devin W. Kolmetzky, John W. Elrod, and Alycia N Hildebrand
- Subjects
biology ,Amyloid beta ,Chemistry ,chemistry.chemical_element ,Calcium ,medicine.disease_cause ,Calcium in biology ,Cell biology ,Extracellular ,biology.protein ,medicine ,Mitochondrial calcium uptake ,Cognitive decline ,Intracellular ,Oxidative stress - Abstract
Alzheimer’s disease (AD) is characterized by the extracellular deposition of amyloid beta, intracellular neurofibrillary tangles, synaptic dysfunction, and neuronal cell death. These phenotypes correlate with and are linked to elevated neuronal intracellular calcium (iCa2+) levels. Recently, our group reported that mitochondrial calcium (mCa2+) overload, due to loss of mCa2+ efflux capacity, contributes to AD development and progression. We also noted proteomic remodeling of the mitochondrial calcium uniporter channel (mtCU) in sporadic AD brain samples, suggestive of altered mCa2+ uptake in AD. Since the mtCU is the primary mechanism for Ca2+ uptake into the mitochondrial matrix, inhibition of the mtCU has the potential to reduce or prevent mCa2+ overload in AD. Here, we report that neuronal-specific loss of mtCU-dependent mCa2+ uptake in the 3xTg-AD mouse model of AD reduced Aβ and tau-pathology, synaptic dysfunction, and cognitive decline. Knockdown of Mcu in a cellular model of AD significantly decreased matrix Ca2+ content, oxidative stress and cell death. These results suggest that inhibition of neuronal mCa2+ uptake is a novel therapeutic target to impede AD progression.
- Published
- 2021
- Full Text
- View/download PDF
32. Abstract 16434: Temporal Metabolomic and Transcriptomic Changes in a Large-Animal Model of Hfpef
- Author
-
Dhanendra Tomar, Anh T Huynh, Deborah M Eaton, Andrew A. Gibb, Remus M. Berretta, Joanne F Garbincius, Devin W. Kolmetzky, Emma K Murray, Steve R. Houser, John W. Elrod, and M. Wallner
- Subjects
medicine.medical_specialty ,business.industry ,Aortic constriction ,medicine.disease ,Transcriptome ,Metabolomics ,Physiology (medical) ,Internal medicine ,Heart failure ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,Heart failure with preserved ejection fraction ,Large animal - Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for ~50% of HF cases, with no effective treatments. We previously reported that a feline aortic banding model recapitulates many of the multi-factorial features of HFpEF, including: LV hypertrophy, left atrial enlargement, elevated LV filling pressures, impaired pulmonary mechanics and fibrosis. Importantly, this model lacks obesity and hypertension enabling the discovery of cardiac centric targets independent of comorbidities. We examined early changes in metabolism and transcription to gain mechanistic insight into HFpEF development. Male short-hair kittens (2mo old) underwent aortic banding or sham operation. Cardiac function was assessed at baseline and 1mo post-banding prior to tissue collection and downstream analyses. Following banding, we observed significant cardiac hypertrophy and initiation of LV fibrosis in the absence of changes in cardiac function. We observed LV mitochondrial dysfunction, indicated by impaired complex-I and -II respiration prompting the examination of cardiac metabolism by unbiased metabolomics. 82 metabolites were significantly different (≥ 1.25 fold, p ≤ 0.1) between 1mo banded and sham hearts, with an overrepresentation of amino acid (aa) and lipid species. Pathway enrichment analysis highlighted an increase in aa metabolism (e.g. serine, proline) that is associated with ECM remodeling and tissue fibrosis. Additionally, an increase in lipid species (i.e. acyl-carnitines) suggests reduced fatty acid utilization and a shift towards glycolysis. Correlations of metabolomics data with mitochondrial function and cardiac phenotyping revealed strong associations between mitochondrial function and the cardiac energy state, as well as aa and LV fibrosis. RNA-seq and enrichment analyses revealed a significant inflammatory response early in disease progression and a decrease in protein/histone acetylation. Collectively, this systems-based approach provides new insights into the cellular biology underlying HFpEF-like disease progression. The metabolic and transcriptional signature that precede the clinical features of HFpEF, will provide new pre-clinical research directions and may yield novel therapeutic targets.
- Published
- 2020
- Full Text
- View/download PDF
33. Abstract 15070: Micu1 Regulates Mitochondrial Cristae Structure and Function Independent of the Mitochondrial Calcium Uniporter Channel
- Author
-
Suresh K. Joseph, Devin W. Kolmetzky, György Hajnóczky, Dhanendra Tomar, April C. Carpenter, Joanne F Garbincius, Manfred Thomas, Pooja Jadiya, John W. Elrod, and Oniel Salik
- Subjects
Cell signaling ,business.industry ,chemistry.chemical_element ,Mitochondrial calcium uniporter ,Mitochondrion ,Calcium ,Cell biology ,Structure and function ,chemistry ,Physiology (medical) ,Mitochondrial cristae ,Medicine ,Cardiology and Cardiovascular Medicine ,Uniporter ,business - Abstract
Background: MICU1 is an EF-hand domain containing Ca 2+ -sensor regulating the mitochondrial Ca 2+ uniporter channel and mitochondrial Ca 2+ uptake. MICU1-null mouse and fly models display perinatal lethality with disorganized mitochondrial architecture. Interestingly, these phenotypes are distinct from other mtCU loss-of-function models ( MCU, MICU2, EMRE, MCUR1 ) and thus are likely not explained solely by changes in matrix Ca 2+ content. Using size-exclusion proteomics and co-immunofluorescence, we found that MICU1 localizes to mitochondrial complexes lacking MCU. These observations suggest that MICU1 may have additional cellular functions independent of the MCU. Methods: Biotin-based proximity labeling and proteomics, protein biochemistry, live-cell Ca 2+ imaging, electron microscopy, confocal and super-resolution imaging were utilized to identify and validate MICU1 novel functions. Results: The expression of a MICU1-BioID2 fusion protein in MCU +/+ and MCU -/- cells allowed the identification of the total vs. MCU-independent MICU1 interactome. LC-MS analysis of purified biotinylated proteins identified the mitochondrial contact site and cristae organizing system (MICOS) components Mitofilin (MIC60) and Coiled-coil-helix-coiled-coil helix domain containing 2 (CHCHD2) as MCU independent novel MICU1 interactors. We demonstrate that MICU1 is essential for proper organization of the MICOS complex and that MICU1 ablation results in altered cristae organization, mitochondrial ultrastructure, mitochondrial membrane dynamics, membrane potential, and cell death signaling. We hypothesize that MICU1 is a MICOS Ca 2+ - sensor since perturbing MICU1 is sufficient to modulate cytochrome c release independent of Ca 2+ uptake across the inner mitochondrial membrane. Conclusions: Here, we provide the first experimental evidence of an intermembrane space Ca 2+ - sensor regulating mitochondrial membrane dynamics, independent of changes in matrix Ca 2+ content. This study provides a novel paradigm to understand Ca 2+ -dependent regulation of mitochondrial structure and function and may help explain the mitochondrial remodeling reported to occur in numerous disease states.
- Published
- 2020
- Full Text
- View/download PDF
34. Abstract 270: Micu1 Regulates Mitochondrial Structural Integrity and Cristae Function Independent of Mcu Calcium Uptake
- Author
-
Devin W. Kolmetzky, April C. Carpenter, Manfred Thomas, Oniel Salik, Joanne F Garbincius, John W. Elrod, Dhanendra Tomar, and Pooja Jadiya
- Subjects
Physiology ,Chemistry ,Structural integrity ,Cardiology and Cardiovascular Medicine ,Calcium uptake ,Function (biology) ,Cell biology - Abstract
MICU1 is a Ca 2+ -sensor regulating the mitochondrial Ca 2+ uniporter channel (mtCU) and mitochondrial Ca 2+ uptake. MICU1 contains two Ca 2+ - binding EF-hand domains and directly interacts with the mtCU pore-forming subunit, MCU. MICU1-null mouse and fly models display perinatal lethality with disorganized mitochondrial architecture. Interestingly, these phenotypes are distinct from other mtCU loss-of-function models ( MCU, MICU2, EMRE, MCUR1 ) and thus are likely not explained solely by changes in matrix Ca 2+ content, suggesting MICU1 has cellular functions independent of the mtCU. To discern novel MICU1 molecular interactions we utilized a proximity-based biotinylation proteomic approach, BioID2, and expressed a MICU1-BioID2 fusion construct in wild-type and MCU -/- cells. LC-MS analysis of purified biotinylated proteins identified Mitochondrial Contact Site and Cristae Organizing System (MICOS) components MIC60, CHCHD2, and CHCHD3 as MICU1 interacting partners. Loss of MCU did not affect the MICU1-MICOS interaction, suggesting that MICU1 could be regulating the MICOS independent of the mtCU. Fast protein liquid chromatography (FPLC), blue native-PAGE, co-immunoprecipitation, live-cell Ca 2+ imaging, electron microscopy, confocal and super-resolution imaging confirmed that MICU1 (independent of matrix Ca 2+ ) is regulating mitochondrial membrane dynamics, membrane potential, and cell death signaling. Further, the deletion of Chchd2, a core component of the MICOS, resulted in disruption of cristae structure without any observable effect on mitochondrial Ca 2+ uptake. In addition, RNA sequencing of murine models of heart failure revealed a correlation between the expression of MICU1 and MICOS components and disease progression. These results are the first to identify how Ca 2+ regulates cristae structure and function and suggests MICU1, at the MICOS, regulates mitochondrial membrane remodeling in the context of cardiac stress and disease independent of the mtCU.
- Published
- 2020
- Full Text
- View/download PDF
35. Abstract 381: Mcur1 is a Potential Target to Modulate Cardiac Contractility and Alleviate Cardiac Function During Heart Failure
- Author
-
Sudarsan Rajan, Santhanam Shanmughapriya, Erhe Gao, Hong Wang, Rajika Roy, Xue-Qian Zhang, Sarah M. Schumacher, Jianliang Song, Dhanendra Tomar, Joseph Y. Cheung, and Walter J. Koch
- Subjects
Cardiac function curve ,Contractility ,medicine.medical_specialty ,Physiology ,business.industry ,Heart failure ,Internal medicine ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business - Abstract
Cardiac contractility is regulated by the intracellular Ca 2+ concentration fluxes which are actively regulated by multiple channels and transporters. Ca 2+ uptake into the mitochondrial matrix is precisely controlled by the highly Ca 2+ selective channel, Mitochondrial Calcium Uniporter (MCU). Earlier studies on the cardiac-specific acute MCU knockout and a transgenic dominant-negative MCU mice have demonstrated that mitochondrial Ca 2+ ( m Ca 2+ ) signaling is necessary for cardiac ‘‘fight-or-flight’’ contractile response, however, the role of m Ca 2+ buffering to shape global cytosolic Ca 2+ levels and affect E-C coupling, particularly the Ca 2+ transient, on a beat-to-beat basis still remains to be solved. Our earlier studies have demonstrated that loss of MCU Regulator 1 (MCUR1) in cardiomyocytes results in the impaired m Ca 2+ uptake. We have now employed the cardiac-specific MCUR1 knockout mouse to dissect the precise role of MCU in regulating cytosolic Ca 2+ transients associated with excitation-contraction (E-C) coupling and cardiac function. Results from our studies including the in vivo analyses of cardiac physiology during normal and pressure-overloaded mouse models and in vitro experiments including single-cell cardiac contractility, calcium transients, and electrophysiology measurements demonstrate that MCUR1/MCU regulated m Ca 2+ buffering in cardiomyocytes, although insignificant under basal condition, becomes critical in stress induced conditions and actively participates in regulating the c Ca 2+ transients. Also, the ablation of MCUR1 in cardiomyocytes during stress conditions prevents m Ca 2+ overload and subsequent mROS overproduction. Our data indicate that MCUR1 ablation offers protection against pressure-overload cardiac hypertrophy. In summary, our results provide critical insights into the mechanisms by which the MCU channel contributes in regulating the contractile function of the cardiomyocytes and the role of m Ca 2+ in the development and progression of heart failure.
- Published
- 2020
- Full Text
- View/download PDF
36. SARS-CoV-2, ACE2, and Hydroxychloroquine: Cardiovascular Complications, Therapeutics, and Clinical Readouts in the Current Settings
- Author
-
Avtar S. Meena, Dhanendra Tomar, Ramesh Kandimalla, and Rajkumar Singh Kalra
- Subjects
Microbiology (medical) ,2019-20 coronavirus outbreak ,medicine.medical_specialty ,hydroxychloroquine ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,ACE2 ,lcsh:Medicine ,Clinical settings ,Review ,therapeutics ,medicine ,Immunology and Allergy ,Medical prescription ,Intensive care medicine ,Molecular Biology ,Repurposing ,General Immunology and Microbiology ,business.industry ,SARS-CoV-2 ,cardiovascular disease (CVD) ,lcsh:R ,COVID-19 ,Hydroxychloroquine ,Infectious Diseases ,cardiovascular system ,business ,medicine.drug ,Healthcare system - Abstract
The rapidly evolving coronavirus disease 2019 (COVID-19, caused by severe acute respiratory syndrome coronavirus 2- SARS-CoV-2), has greatly burdened the global healthcare system and led it into crisis in several countries. Lack of targeted therapeutics led to the idea of repurposing broad-spectrum drugs for viral intervention. In vitro analyses of hydroxychloroquine (HCQ)’s anecdotal benefits prompted its widespread clinical repurposing globally. Reports of emerging cardiovascular complications due to its clinical prescription are revealing the crucial role of angiotensin-converting enzyme 2 (ACE2), which serves as a target receptor for SARS-CoV-2. In the present settings, a clear understanding of these targets, their functional aspects and physiological impact on cardiovascular function are critical. In an up-to-date format, we shed light on HCQ’s anecdotal function in stalling SARS-CoV-2 replication and immunomodulatory activities. While starting with the crucial role of ACE2, we here discuss the impact of HCQ on systemic cardiovascular function, its associated risks, and the scope of HCQ-based regimes in current clinical settings. Citing the extent of HCQ efficacy, the key considerations and recommendations for the use of HCQ in clinics are further discussed. Taken together, this review provides crucial insights into the role of ACE2 in SARS-CoV-2-led cardiovascular activity, and concurrently assesses the efficacy of HCQ in contemporary clinical settings.
- Published
- 2020
37. Mitochondrial Protein Quality Control Mechanisms
- Author
-
Pooja Jadiya and Dhanendra Tomar
- Subjects
0301 basic medicine ,Cell signaling ,Proteasome Endopeptidase Complex ,Protein Folding ,lcsh:QH426-470 ,Bioenergetics ,proteome ,Review ,Mitochondrion ,Biology ,Genome ,Mitochondrial Proteins ,03 medical and health sciences ,0302 clinical medicine ,Mitochondrial unfolded protein response ,Mitophagy ,Genetics ,Humans ,chaperones ,mitochondria-associated degradation ,Genetics (clinical) ,mitochondrial protein quality control ,Cell Nucleus ,Ubiquitin ,protease ,Cell biology ,Mitochondria ,lcsh:Genetics ,Cytosol ,030104 developmental biology ,proteasome ,Protein Biosynthesis ,mitochondrial unfolded protein response ,Proteome ,Unfolded Protein Response ,030217 neurology & neurosurgery - Abstract
Mitochondria serve as a hub for many cellular processes, including bioenergetics, metabolism, cellular signaling, redox balance, calcium homeostasis, and cell death. The mitochondrial proteome includes over a thousand proteins, encoded by both the mitochondrial and nuclear genomes. The majority (~99%) of proteins are nuclear encoded that are synthesized in the cytosol and subsequently imported into the mitochondria. Within the mitochondria, polypeptides fold and assemble into their native functional form. Mitochondria health and integrity depend on correct protein import, folding, and regulated turnover termed as mitochondrial protein quality control (MPQC). Failure to maintain these processes can cause mitochondrial dysfunction that leads to various pathophysiological outcomes and the commencement of diseases. Here, we summarize the current knowledge about the role of different MPQC regulatory systems such as mitochondrial chaperones, proteases, the ubiquitin-proteasome system, mitochondrial unfolded protein response, mitophagy, and mitochondria-derived vesicles in the maintenance of mitochondrial proteome and health. The proper understanding of mitochondrial protein quality control mechanisms will provide relevant insights to treat multiple human diseases.
- Published
- 2020
38. Mitochondrial dysfunction in human immunodeficiency virus‐1 transgenic mouse cardiac myocytes
- Author
-
Fabian Jana Prado, Nana Merabova, Manish K. Gupta, Joseph M. McClung, Joseph Y. Cheung, Jennifer Gordon, Jianliang Song, Xue-Qian Zhang, Santhanam Shanmughapriya, Sudarsan Rajan, Kamel Khalili, Christopher D. Kontos, Tijana Knezevic, Muniswamy Madesh, Dhanendra Tomar, Arthur M. Feldman, Farzaneh G. Tahrir, Paul E. Klotman, and JuFang Wang
- Subjects
0301 basic medicine ,Membrane potential ,chemistry.chemical_classification ,Genetically modified mouse ,Reactive oxygen species ,Contraction (grammar) ,Physiology ,Chemistry ,Protein subunit ,Transgene ,Clinical Biochemistry ,Wild type ,Cell Biology ,Molecular biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Myocyte - Abstract
The pathophysiology of human immunodeficiency virus (HIV)-associated cardiomyopathy remains uncertain. We used HIV-1 transgenic (Tg26) mice to explore mechanisms by which HIV-related proteins impacted on myocyte function. Compared to adult ventricular myocytes isolated from nontransgenic (wild type [WT]) littermates, Tg26 myocytes had similar mitochondrial membrane potential (ΔΨ m ) under normoxic conditions but lower Δ Ψ m after hypoxia/reoxygenation (H/R). In addition, Δ Ψ m in Tg26 myocytes failed to recover after Ca 2+ challenge. Functionally, mitochondrial Ca 2+ uptake was severely impaired in Tg26 myocytes. Basal and maximal oxygen consumption rates (OCR) were lower in normoxic Tg26 myocytes, and further reduced after H/R. Complex I subunit and ATP levels were lower in Tg26 hearts. Post-H/R, mitochondrial superoxide (O 2 •- ) levels were higher in Tg26 compared to WT myocytes. Overexpression of B-cell lymphoma 2-associated athanogene 3 (BAG3) reduced O 2 •- levels in hypoxic WT and Tg26 myocytes back to normal. Under normoxic conditions, single myocyte contraction dynamics were similar between WT and Tg26 myocytes. Post-H/R and in the presence of isoproterenol, myocyte contraction amplitudes were lower in Tg26 myocytes. BAG3 overexpression restored Tg26 myocyte contraction amplitudes to those measured in WT myocytes post-H/R. Coimmunoprecipitation experiments demonstrated physical association of BAG3 and the HIV protein Tat. We conclude: (a) Under basal conditions, mitochondrial Ca 2+ uptake, OCR, and ATP levels were lower in Tg26 myocytes; (b) post-H/R, Δ Ψ m was lower, mitochondrial O 2 •- levels were higher, and contraction amplitudes were reduced in Tg26 myocytes; and (c) BAG3 overexpression decreased O 2 •- levels and restored contraction amplitudes to normal in Tg26 myocytes post-H/R in the presence of isoproterenol.
- Published
- 2018
- Full Text
- View/download PDF
39. Egr-1 mediated cardiac miR-99 family expression diverges physiological hypertrophy from pathological hypertrophy
- Author
-
Tharmarajan Ramprasath, Dhanendra Tomar, Suresh Shanmugarajan, Karuppusamy V. Karthik, K. Shanmugha Rajan, Ganesan Velmurugan, Pandi Gopal, Subbiah Ramasamy, Suresh K Verma, Balakrishnan Rekha, Rajan Sudarsan, Sivakumar Anusha, and Venkata Naga Srikanth Garikipati
- Subjects
0301 basic medicine ,Programmed cell death ,Small RNA ,Down-Regulation ,Cardiomegaly ,030204 cardiovascular system & hematology ,Biology ,Cell Line ,Muscle hypertrophy ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,microRNA ,Animals ,Gene silencing ,Myocyte ,Myocytes, Cardiac ,Rats, Wistar ,Promoter Regions, Genetic ,Transcription factor ,Early Growth Response Protein 1 ,Cell Biology ,Rats ,Up-Regulation ,Cell biology ,MicroRNAs ,030104 developmental biology ,Signal Transduction - Abstract
The physiological cardiac hypertrophy is an adaptive condition without myocyte cell death, while pathological hypertrophy is a maladaptive condition associated with myocyte cell death. This study explores the miRNome of α-2M-induced physiologically hypertrophied cardiomyocytes and the role of miRNA-99 family during cardiac hypertrophy. Physiological and pathological cardiac hypertrophy was induced in H9c2 cardiomyoblast cell lines using α-2M and isoproterenol respectively. Total RNA isolation and small RNA sequencing were executed for physiological hypertrophy model. The differentially expressed miRNAs and its target mRNAs were validated in animal models. Transcription factor binding sites were predicted in the promoter of specific miRNAs and validated by ChIP-PCR. Subsequently, the selected miRNA was functionally characterized by overexpression and silencing. The effects of silencing of upstream regulator and downstream target gene were studied. Analysis of small RNA reads revealed the differential expression of a large set of miRNAs during hypertrophy, of which miR-99 family was highly downregulated upon α-2M treatment. However, this miR-99 family expression was upregulated during pathological hypertrophy and confirmed in animal models. ChIP-PCR confirms the binding of Egr-1 transcription factor to the miR-99 promoter. Further, silencing of Egr-1 decreased the expression of miR-99. The overexpression or silencing of miR-99 diverges the physiological hypertrophy to pathological hypertrophy and vice versa by regulating Akt-1 pathway. Silencing of Akt-1 replicates the effect of overexpression of miR-99. Conclusion The results proved Egr-1 mediated regulation of miR-99 family that plays a key role in determining the fate of cardiac hypertrophy by regulating Akt-1 signaling.
- Published
- 2018
- Full Text
- View/download PDF
40. MIRO-1 Determines Mitochondrial Shape Transition upon GPCR Activation and Ca2+ Stress
- Author
-
Santhanam Shanmughapriya, Andrea Ketschek, William J. Craigen, Julie Heffler, Sudarsan Rajan, Fabián Jaña, David C. Chan, Aparna Tripathi, Ramasamy Subbiah, Benjamin L. Prosser, Massimo F. Riitano, Edmund Carvalho, Shuxia Meng, Jeffrey L. Caplan, Alison M. Worth, Hiromi Sesaki, Palaniappan Palaniappan, Zhiwei Dong, Muniswamy Madesh, Dhanendra Tomar, Thomas Manfred, Neeharika Nemani, Gianluca Gallo, Kie Itoh, Prashant Mishra, Sarah L. Breves, Ajay Seelam, Donald L. Gill, and Peter B. Stathopulos
- Subjects
0301 basic medicine ,Programmed cell death ,Tubulin complex ,EF hand ,Chemistry ,chemistry.chemical_element ,Mitochondrion ,Calcium ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cytosol ,030104 developmental biology ,0302 clinical medicine ,lcsh:Biology (General) ,Mitophagy ,Biophysics ,otorhinolaryngologic diseases ,Mitochondrial fission ,lcsh:QH301-705.5 ,030217 neurology & neurosurgery - Abstract
Summary: Mitochondria shape cytosolic calcium ([Ca2+]c) transients and utilize the mitochondrial Ca2+ ([Ca2+]m) in exchange for bioenergetics output. Conversely, dysregulated [Ca2+]c causes [Ca2+]m overload and induces permeability transition pore and cell death. Ablation of MCU-mediated Ca2+ uptake exhibited elevated [Ca2+]c and failed to prevent stress-induced cell death. The mechanisms for these effects remain elusive. Here, we report that mitochondria undergo a cytosolic Ca2+-induced shape change that is distinct from mitochondrial fission and swelling. [Ca2+]c elevation, but not MCU-mediated Ca2+ uptake, appears to be essential for the process we term mitochondrial shape transition (MiST). MiST is mediated by the mitochondrial protein Miro1 through its EF-hand domain 1 in multiple cell types. Moreover, Ca2+-dependent disruption of Miro1/KIF5B/tubulin complex is determined by Miro1 EF1 domain. Functionally, Miro1-dependent MiST is essential for autophagy/mitophagy that is attenuated in Miro1 EF1 mutants. Thus, Miro1 is a cytosolic Ca2+ sensor that decodes metazoan Ca2+ signals as MiST. : Metazoan Ca2+ signal determines mitochondrial shape transition (MiST) and cellular quality control. Nemani et al. find that mitochondria undergo shape changes upon Ca2+ stress. MiST is distinct from matrix Ca2+-induced swelling and mitochondrial dynamics. The conserved Ca2+ sensor Miro1 enables MiST and promotes autophagy/mitophagy. Keywords: mitochondrial shape, MiST, calcium, Miro, EF hand, PTP, MCU, mitophagy, autophagy, mitochondrial dynamics
- Published
- 2018
41. Calcium and Bioenergetics
- Author
-
Pooja Jadiya and Dhanendra Tomar
- Subjects
Programmed cell death ,chemistry ,Bioenergetics ,chemistry.chemical_element ,Mitochondrial calcium uniporter ,Metabolism ,Calcium ,Mitochondrion ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
42. Platelet microparticles infiltrating solid tumors transfer miRNAs that suppress tumor growth
- Author
-
Nicholas E. Hoffman, Sudarsan Rajan, Lawrence E. Goldfinger, Dhanendra Tomar, Mikhail A. Kolpakov, Abdelkarim Sabri, Johnny Yu, Leonard C. Edelstein, Guang Fen Mao, James V. Michael, Jeremy G.T. Wurtzel, Andrew S. Weyrich, Marvin T. Nieman, A. Koneti Rao, Jesse W. Rowley, and Muniswamy Madesh
- Subjects
Blood Platelets ,0301 basic medicine ,Lung Neoplasms ,Receptors, Proteinase-Activated ,Immunology ,Cell ,Biology ,Biochemistry ,Article ,Metastasis ,Mice ,03 medical and health sciences ,Cell-Derived Microparticles ,In vivo ,Gene expression ,microRNA ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Small nucleolar RNA ,RNA ,NADH Dehydrogenase ,Cell Biology ,Hematology ,medicine.disease ,Molecular biology ,Neoplasm Proteins ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,Colonic Neoplasms ,Cancer research - Abstract
Platelet-derived microparticles (PMPs) are associated with enhancement of metastasis and poor cancer outcomes. Circulating PMPs transfer platelet microRNAs (miRNAs) to vascular cells. Solid tumor vasculature is highly permeable, allowing the possibility of PMP-tumor cell interaction. Here, we show that PMPs infiltrate solid tumors in humans and mice and transfer platelet-derived RNA, including miRNAs, to tumor cells in vivo and in vitro, resulting in tumor cell apoptosis. MiR-24 was a major species in this transfer. PMP transfusion inhibited growth of both lung and colon carcinoma ectopic tumors, whereas blockade of miR-24 in tumor cells accelerated tumor growth in vivo, and prevented tumor growth inhibition by PMPs. Conversely, Par4-deleted mice, which had reduced circulating microparticles (MPs), supported accelerated tumor growth which was halted by PMP transfusion. PMP targeting was associated with tumor cell apoptosis in vivo. We identified direct RNA targets of platelet-derived miR-24 in tumor cells, which included mitochondrial mt-Nd2, and Snora75, a noncoding small nucleolar RNA. These RNAs were suppressed in PMP-treated tumor cells, resulting in mitochondrial dysfunction and growth inhibition, in an miR-24-dependent manner. Thus, platelet-derived miRNAs transfer in vivo to tumor cells in solid tumors via infiltrating MPs, regulate tumor cell gene expression, and modulate tumor progression. These findings provide novel insight into mechanisms of horizontal RNA transfer and add multiple layers to the regulatory roles of miRNAs and PMPs in tumor progression. Plasma MP-mediated transfer of regulatory RNAs and modulation of gene expression may be a common feature with important outcomes in contexts of enhanced vascular permeability.
- Published
- 2017
- Full Text
- View/download PDF
43. MICU1 regulates mitochondrial cristae structure and function independent of the mitochondrial calcium uniporter channel
- Author
-
Manfred Thomas, Devin W. Kolmetzky, April C. Carpenter, Pooja Jadiya, Oniel Salik, John W. Elrod, Dhanendra Tomar, and Joanne F Garbincius
- Subjects
0303 health sciences ,Chemistry ,Protein subunit ,Cellular functions ,Mitochondrial calcium uniporter ,Gating ,Proteomics ,Structure and function ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Mitochondrial cristae ,Uniporter ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
MICU1 is an EF-hand-containing mitochondrial protein that is essential for gating of the mitochondrial Ca2+ uniporter channel (mtCU) and is reported to interact directly with the pore-forming subunit, MCU and scaffold EMRE. However, using size-exclusion proteomics, we found that MICU1 exists in mitochondrial complexes lacking MCU. This suggests that MICU1 may have additional cellular functions independent of regulating mitochondrial Ca2+ uptake. To discern mtCU-independent MICU1 functions, we employed a proteomic discovery approach using BioID2-mediated proximity-based (MICU1-/- and MCU-/- cells allowed the identification of total vs. mtCU-independent MICU1 interactors. Bioinformatics identified the Mitochondrial Contact Site and Cristae Organizing System (MICOS) components MIC60 (encoded by the IMMT gene) and Coiled-coil-helix-coiled-coil helix domain containing 2 (CHCHD2) as novel MICU1 interactors, independent of the mtCU. We demonstrate that MICU1 is essential for proper proteomic organization of the MICOS complex and that MICU1 ablation results in altered cristae organization and mitochondrial ultrastructure. We hypothesize that MICU1 serves as a MICOS calcium sensor, since perturbing MICU1 is sufficient to modulate cytochrome c release independent of mitochondrial Ca2+ uptake across the inner mitochondrial membrane (IMM). Here, we provide the first experimental evidence suggesting that MICU1 regulates cellular functions independent of mitochondrial calcium uptake and may serve as a critical mediator of Ca2+-dependent signaling to modulate mitochondrial membrane dynamics and cristae organization.
- Published
- 2019
- Full Text
- View/download PDF
44. Abstract 101: Identification of Novel MICU1 Interactors Independent of the mtCU Complex
- Author
-
Joanne F Garbincius, John W. Elrod, Manfred Thomas, Devin W. Kolmetzky, Dhanendra Tomar, and Pooja Jadiya
- Subjects
Physiology ,Chemistry ,Protein subunit ,Cardiac metabolism ,chemistry.chemical_element ,Identification (biology) ,Calcium ,Mitochondrion ,Cardiology and Cardiovascular Medicine ,Uniporter ,Mitochondrial protein ,Cell biology - Abstract
MICU1 is an EF-hand containing mitochondrial protein that gates the mitochondrial Ca 2+ uniporter complex (mtCU) and directly interacts with the pore-forming subunit MCU. Previous studies have shown perinatal lethality and altered mitochondrial architecture in MICU1 knockout ( Micu1 -/- ) mice, phenotypes that are distinct from other knockout models of mtCU components, such as MCU, and thus are likely not explained solely by changes in matrix [Ca 2+ ] uptake. Further, our proteomic studies suggest that MICU1 exists in mitochondrial complexes void of MCU. This suggests that MICU1 may have cellular functions independent of mtCU regulation. To discern novel MICU1 molecular interactors we employed a biotinylation-based proteomic approach in Mcu -/- and Micu1 -/- cells to detect proteins interacting with MICU1 using fusion protein containing BioID2, a small biotin ligase for proximity-dependent labeling. Expression of MICU1-BioID2 in Mcu -/- cells allowed the identification of mtCU-independent interactors. Fast protein liquid chromatography (FPLC), blue native-PAGE, co-immunoprecipitation, live-cell Ca 2+ imaging, confocal and super-resolution imaging methods were used to confirm novel roles for MICU1 in mitochondrial biology. LC-MS analysis of biotinylated proteins after avidin-based purification identified the Mitochondrial Contact Site and Cristae Organizing System (MICOS) components IMMT, CHCHD2, and CHCHD3 as interacting with MICU1 (MICU1-BioD2 expressed in Micu1 -/- cells to avoid aberrant expression). These same MICOS components were identified in MCU -/- cells, suggesting that MICU1 could be involved in the regulation of MICOS independent of the mtCU. Further, the deletion of CHCHD2 resulted in the loss of MICOS and cristae disorganization without any observable effect on m Ca 2+ uptake. RNA sequencing revealed correlative expression changes in MICU1 and MICOS components in response to heart failure progression during transverse aortic constriction and myocardial infarction. These results suggest MICU1 likely serves cellular functions independent of the mtCU and may serve as a key regulator of Ca 2+ -dependent signaling (EF-hands) in other cellular processes that are dysregulated during heart failure.
- Published
- 2019
- Full Text
- View/download PDF
45. Abstract 857: MCUB Regulates the Molecular Composition of the Mitochondrial Calcium Uniporter Channel During Cardiac Stress to Limit Mitochondrial Calcium Overload
- Author
-
Erhe Gao, John W. Elrod, Timothy S. Luongo, Anna Maria Lucchese, Dhanendra Tomar, Pooja Jadiya, Xue-Qian Zhang, Devin W. Kolmetzky, Neil Shah, and Jonathan P. Lambert
- Subjects
Molecular composition ,chemistry ,Physiology ,chemistry.chemical_element ,Mitochondrial calcium uniporter ,Ischemic injury ,Calcium ,Mitochondrion ,Cardiology and Cardiovascular Medicine ,Calcium overload ,Inner mitochondrial membrane ,Cell biology - Abstract
The mitochondrial calcium uniporter (mtCU) is a ~700 kD multi-subunit channel residing in the inner mitochondrial membrane required for mitochondrial Ca 2+ ( m Ca 2+ ) uptake. Mitochondrial Calcium Uniporter B ( MCUB ) is reported to negatively regulate m Ca 2+ uptake, but its precise functional role and contribution to cardiac physiology remain unresolved. Size exclusion chromatography of ventricular mitochondria revealed MCUB was absent from high-molecular weight (MW) mtCU complexes in sham animals, but present 24 hours following myocardial ischemia-reperfusion injury (IR). To investigate MCUB ’s contribution to mtCU regulation we created a MCUB -/- cell line by CRISPR-Cas9n. MCUB deletion increased histamine-mediated [ m Ca 2+ ] transient amplitude by ~50% vs. WT controls (mito-R-GECO1). MCUB deletion increased mtCU capacitance (mitoplast patch-clamp) and rate of [ m Ca 2+ ] uptake. Size-exclusion chromatography revealed loss of MCUB increased MCU incorporation into high-MW mtCU, suggesting stoichiometric replacement and overall more functional mtCU’s. To examine MCUB’s role in cardiac physiology we generated a cardiac-specific, tamoxifen-inducible MCUB mouse model (CAG-CAT-MCUB x MCM; MCUB-Tg). FPLC revealed MCUB was undetected in high-MW mtCU complexes of Cre controls, but enriched in MCUB-Tg hearts. MCUB incorporation decreased the presence of channel gatekeepers, MICU1/2, and decreased the MW of the mtCU complex. Immunoprecipitations suggest MCUB interacts with MCU but not MICU1/2. MCUB-Tg adult cardiomyocytes (ACMs) expressing AAV9-mitycam ( m Ca 2+ reporter) were paced and displayed a ~30% decrease in m Ca 2+ transient peak amplitude with significantly reduced m Ca 2+ uptake rates vs controls. A reduction in OxPhos reserve capacity correlated with a severe impairment in cardiac contractile reserve (LV invasive hemodynamics during isoproterenol infusion). MCUB-Tg cardiac mitochondria were resistant to Ca 2+ -induced permeability transition and MCUB-Tg mice displayed ~50% decrease in infarct size per area-at-risk after in vivo IR-injury. These data suggest MCUB regulation of the mtCU is an endogenous compensatory mechanism to decrease m Ca 2+ overload during ischemic injury, but maladaptive to cardiac energetic responsiveness.
- Published
- 2019
- Full Text
- View/download PDF
46. The relationship between DJ-1 and S100A8 in human primary alveolar type II cells in emphysema
- Author
-
Chih-Ru Lin, Nathaniel Marchetti, Karim Bahmed, Mark A. Wilson, Beata Kosmider, Sudhir Bolla, Dhanendra Tomar, Muniswamy Madesh, and Gerard J. Criner
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Male ,Programmed cell death ,Pathology ,medicine.medical_specialty ,Physiology ,Pulmonary emphysema ,Protein Deglycase DJ-1 ,Apoptosis ,medicine.disease_cause ,S100A8 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Physiology (medical) ,Smoke ,medicine ,Cigarette smoke ,Animals ,Humans ,Calgranulin A ,Aged ,Alveolar Wall ,Mice, Knockout ,Alveolar type ,business.industry ,Cell Biology ,respiratory system ,Middle Aged ,Mice, Inbred C57BL ,Pulmonary Alveoli ,Oxidative Stress ,030104 developmental biology ,Pulmonary Emphysema ,Cytoprotection ,030220 oncology & carcinogenesis ,Alveolar Epithelial Cells ,Female ,business ,Oxidative stress ,Research Article - Abstract
Pulmonary emphysema is characterized by alveolar type II (ATII) cell death, destruction of alveolar wall septa, and irreversible airflow limitation. Cigarette smoke induces oxidative stress and is the main risk factor for this disease development. ATII cells isolated from nonsmokers, smokers, and patients with emphysema were used for this study. ATII cell apoptosis in individuals with this disease was detected. DJ-1 and S100A8 have cytoprotective functions against oxidative stress-induced cell injury. Reduced DJ-1 and S100A8 interaction was found in ATII cells in patients with emphysema. The molecular function of S100A8 was determined by an analysis of the oxidation status of its cysteine residues using chemoselective probes. Decreased S100A8 sulfination was observed in emphysema patients. In addition, its lower levels correlated with higher cell apoptosis induced by cigarette smoke extract in vitro. Cysteine at position 106 within DJ-1 is a central redox-sensitive residue. DJ-1 C106A mutant construct abolished the cytoprotective activity of DJ-1 against cell injury induced by cigarette smoke extract. Furthermore, a molecular and complementary relationship between DJ-1 and S100A8 was detected using gain- and loss-of-function studies. DJ-1 knockdown sensitized cells to apoptosis induced by cigarette smoke extract, and S100A8 overexpression provided cytoprotection in the absence of DJ-1. DJ-1 knockout mice were more susceptible to ATII cell apoptosis induced by cigarette smoke compared with wild-type mice. Our results indicate that the impairment of DJ-1 and S100A8 function may contribute to cigarette smoke-induced ATII cell injury and emphysema pathogenesis.
- Published
- 2019
47. The central role of protein kinase C epsilon in cyanide cardiotoxicity and its treatment
- Author
-
Salim Merali, Philippe Haouzi, Annick Judenherc-Haouzi, Jianliang Song, Joseph Y. Cheung, Hanning You, Dhanendra Tomar, JuFang Wang, Xue-Qian Zhang, and Carmen Merali
- Subjects
0301 basic medicine ,Calcium channel ,Protein Kinase C-epsilon ,030204 cardiovascular system & hematology ,Toxicology ,Contractility ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Organ Specific Toxicology ,chemistry ,Biophysics ,Phosphorylation ,Kinase activity ,Sodium cyanide ,Protein kinase C ,Intracellular - Abstract
In adult mouse myocytes, brief exposure to sodium cyanide (CN) in the presence of glucose does not decrease ATP levels, yet produces profound reduction in contractility, intracellular Ca2+ concentration ([Ca2+]i) transient and L-type Ca2+ current (ICa) amplitudes. We analyzed proteomes from myocytes exposed to CN, focusing on ionic currents associated with excitation-contraction coupling. CN induced phosphorylation of α1c subunit of L-type Ca2+ channel and α2 subunit of Na+-K+-ATPase. Methylene blue (MB), a CN antidote that we previously reported to ameliorate CN-induced reduction in contraction, [Ca2+]i transient and ICa amplitudes, was able to reverse this phosphorylation. CN decreased Na+-K+-ATPase current contributed by α2 but not α1 subunit, an effect that was also counteracted by MB. Peptide consensus sequences suggested CN-induced phosphorylation was mediated by protein kinase C epsilon (PKCε). Indeed, CN stimulated PKC kinase activity and induced PKCε membrane translocation, effects that were prevented by MB. Pretreatment with myristoylated PKCε translocation activator or inhibitor peptides mimicked and inhibited the effects of CN on ICa and myocyte contraction, respectively. We conclude that CN activates PKCε, which phosphorylates L-type Ca2+ channel and Na+-K+-ATPase, resulting in depressed cardiac contractility. We hypothesize that this inhibition of ion fluxes represents a novel mechanism by which the cardiomyocyte reduces its ATP demand (decreased ion fluxes and contractility), diminishes ATP turnover and preserves cell viability. However, this cellular protective effect translates into life-threatening cardiogenic shock in vivo, thereby creating a profound disconnect between survival mechanisms at the cardiomyocyte level from those at the level of the whole organism.
- Published
- 2019
48. Chemically synthesized Secoisolariciresinol diglucoside (LGM2605) improves mitochondrial function in cardiac myocytes and alleviates septic cardiomyopathy
- Author
-
Hong Wang, Charikleia Kalliora, Anastasios Lymperopoulos, Anna Maria Lucchese, Dimitra Kokkinaki, Dhanendra Tomar, Jennifer Maning, Ioannis D. Kyriazis, Xiaofeng Yang, Melpo Christofidou-Solomidou, Walter J. Koch, Konstantinos Drosatos, Muniswamy Madesh, Santhanam Shanmughapriya, Matthew Hoffman, and Joon-Young Park
- Subjects
0301 basic medicine ,Cardiac function curve ,Punctures ,030204 cardiovascular system & hematology ,Mitochondrion ,Pharmacology ,Article ,Antioxidants ,Mitochondria, Heart ,Cell Line ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,Oxygen Consumption ,Glucosides ,Autophagy ,Medicine ,Animals ,Humans ,Mitochondrial calcium uptake ,Myocytes, Cardiac ,Butylene Glycols ,Molecular Biology ,Cecum ,Ligation ,Heart metabolism ,Membrane Potential, Mitochondrial ,Organelle Biogenesis ,business.industry ,Septic shock ,Myocardium ,Organ dysfunction ,NF-kappa B ,Hypothermia ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,Gene Expression Regulation ,Cytokines ,Calcium ,medicine.symptom ,Inflammation Mediators ,Cardiology and Cardiovascular Medicine ,business ,Cardiomyopathies ,Biomarkers - Abstract
Sepsis is the overwhelming systemic immune response to infection, which can result in multiple organ dysfunction and septic shock. Myocardial dysfunction during sepsis is associated with advanced disease and significantly increased in-hospital mortality. Our group has shown that energetic failure and excess reactive oxygen species (ROS) generation constitute major components of myocardial dysfunction in sepsis. Because ROS production is central to cellular metabolic health, we tested if the synthetic anti-oxidant lignan secoisolariciresinol diglucoside (SDG; LGM2605) would alleviate septic cardiac dysfunction and investigated the underlying mechanism. Using the cecal ligation and puncture (CLP) mouse model of peritonitis-induced sepsis, we observed impairment of cardiac function beginning at 4 h post-CLP surgery. Treatment of mice with LGM2605 (100 mg/kg body weight, i.p.) 6 h post-CLP surgery reduced cardiac ROS accumulation and restored cardiac function. Assessment of mitochondrial respiration (Seahorse XF) in primary cardiomyocytes obtained from adult C57BL/6 mice that had undergone CLP and treatment with LGM2605 showed restored basal and maximal respiration, as well as preserved oxygen consumption rate (OCR) associated with spare capacity. Further analyses aiming to identify the cellular mechanisms that may account for improved cardiac function showed that LGM2605 restored mitochondria abundance, increased mitochondrial calcium uptake and preserved mitochondrial membrane potential. In addition to protecting against cardiac dysfunction, daily treatment with LGM2605 and antibiotic ertapenem (70 mg/kg) protected against CLP-associated mortality and reversed hypothermia when compared against mice receiving ertapenem and saline. Therefore, treatment of septic mice with LGM2605 emerges as a novel pharmacological approach that reduces cardiac ROS accumulation, protects cardiac mitochondrial function, alleviates cardiac dysfunction, and improves survival.
- Published
- 2019
49. Small conductance calcium-activated potassium current and the mechanism of atrial arrhythmia in mice with dysfunctional melanocyte-like cells
- Author
-
Wei-Chung Tsai, Vickas V. Patel, Maria Kudela, Thomas H. Everett, Peng Sheng Chen, Po-Cheng Chang, Zhenhui Chen, Dhanendra Tomar, Changyu Shen, Yi Hsin Chan, Chia Hsiang Hsueh, Eue Keun Choi, Nicholas Anzalone, Michael A. Olaopa, Michael Rubart-von der Lohe, Shien-Fong Lin, Pooja Jadiya, and Emily Luvison
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Small-Conductance Calcium-Activated Potassium Channels ,Statistics as Topic ,chemistry.chemical_element ,030204 cardiovascular system & hematology ,Melanocyte ,Calcium ,Apamin ,Bioinformatics ,Article ,Membrane Potentials ,SK channel ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,SK3 ,Heart Conduction System ,Physiology (medical) ,Internal medicine ,Atrial Fibrillation ,medicine ,Animals ,Heart Atria ,Melanins ,Membrane potential ,business.industry ,Conductance ,Voltage-Sensitive Dye Imaging ,Up-Regulation ,Intramolecular Oxidoreductases ,body regions ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Cardiology and Cardiovascular Medicine ,business ,Dopachrome tautomerase - Abstract
Background The melanin synthesis enzyme dopachrome tautomerase (Dct) regulates intracellular Ca 2+ in melanocytes. Homozygous Dct knockout (Dct −/− ) adult mice are vulnerable to atrial arrhythmias (AA). Objective The purpose of this study was to determine whether apamin-sensitive small conductance Ca 2+ -activated K + (SK) currents are upregulated in Dct −/− mice and contribute to AA. Methods Optical mapping was used to study the membrane potential of the right atrium in Langendorff perfused Dct −/− (n = 9) and Dct +/− (n = 9) mice. Results Apamin prolonged action potential duration (APD) by 18.8 ms (95% confidence interval [CI] 13.4–24.1 ms) in Dct −/− mice and by 11.5 ms (95% CI 5.4–17.6 ms) in Dct +/− mice at a pacing cycle length of 150 ms ( P = .047). The pacing cycle length threshold to induce APD alternans was 48 ms (95% CI 34–62 ms) for Dct −/− mice and 21 ms (95% CI 12–29 ms) for Dct +/− mice ( P = .002) at baseline, and it was 35 ms (95% CI 21–49 ms) for Dct −/− mice and 22 ms (95% CI 11–32 ms) for Dct +/− mice ( P = .025) after apamin administration. Apamin prolonged post-burst pacing APD by 8.9 ms (95% CI 3.9–14.0 ms) in Dct −/− mice and by 1.5 ms (95% CI 0.7–2.3 ms) in Dct +/− mice ( P = .005). Immunoblot and quantitative polymerase chain reaction analyses showed that protein and transcripts levels of SK1 and SK3 were increased in the right atrium of Dct −/− mice. AA inducibility (89% vs 11%; P = .003) and duration (281 seconds vs 66 seconds; P = .008) were greater in Dct −/− mice than in Dct +/− mice at baseline, but not different (22% vs 11%; P = 1.00) after apamin administration. Five of 8 (63%) induced atrial fibrillation episodes in Dct −/− mice had focal drivers. Conclusion Apamin-sensitive SK current upregulation in Dct −/− mice plays an important role in the mechanism of AA.
- Published
- 2016
- Full Text
- View/download PDF
50. BAG3 regulates contractility and Ca2+ homeostasis in adult mouse ventricular myocytes
- Author
-
Arthur M. Feldman, JuFang Wang, Valerie D. Myers, Jianliang Song, Xue-Qian Zhang, Kamel Khalili, Douglas G. Tilley, Feifei Su, Walter J. Koch, Jennifer Gordon, Joseph E. Rabinowitz, Joseph Y. Cheung, Erhe Gao, Muniswamy Madesh, Nicholas E. Hoffman, and Dhanendra Tomar
- Subjects
Cardiomyopathy, Dilated ,0301 basic medicine ,medicine.medical_specialty ,Calcium Channels, L-Type ,Heart Ventricles ,Phospholemman ,Action Potentials ,030204 cardiovascular system & hematology ,Biology ,Article ,Contractility ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Sarcolemma ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Homeostasis ,Humans ,Myocyte ,Myocytes, Cardiac ,RNA, Small Interfering ,Molecular Biology ,Excitation Contraction Coupling ,Adaptor Proteins, Signal Transducing ,Heart Failure ,Membrane potential ,Forskolin ,Voltage-dependent calcium channel ,Endoplasmic reticulum ,Isoproterenol ,Membrane Proteins ,Arrhythmias, Cardiac ,Phosphoproteins ,030104 developmental biology ,Endocrinology ,chemistry ,Calcium ,Receptors, Adrenergic, beta-1 ,Sodium-Potassium-Exchanging ATPase ,Apoptosis Regulatory Proteins ,Cardiology and Cardiovascular Medicine - Abstract
Bcl2-associated athanogene 3 (BAG3) is a 575 amino acid anti-apoptotic protein that is constitutively expressed in the heart. BAG3 mutations, including mutations leading to loss of protein, are associated with familial cardiomyopathy. Furthermore, BAG3 levels have been found to be reduced in end-stage non-familial failing myocardium. In contrast to neonatal myocytes in which BAG3 is found in the cytoplasm and involved in protein quality control and apoptosis, in adult mouse left ventricular (LV) myocytes BAG3 co-localized with Na(+)-K(+)-ATPase and L-type Ca(2+) channels in the sarcolemma and t-tubules. BAG3 co-immunoprecipitated with β1-adrenergic receptor, L-type Ca(2+) channels and phospholemman. To simulate decreased BAG3 protein levels observed in human heart failure, we targeted BAG3 by shRNA (shBAG3) in adult LV myocytes. Reducing BAG3 by 55% resulted in reduced contraction and [Ca(2+)]i transient amplitudes in LV myocytes stimulated with isoproterenol. L-type Ca(2+) current (ICa) and sarcoplasmic reticulum (SR) Ca(2+) content but not Na(+)/Ca(2+) exchange current (INaCa) or SR Ca(2+) uptake were reduced in isoproterenol-treated shBAG3 myocytes. Forskolin or dibutyryl cAMP restored ICa amplitude in shBAG3 myocytes to that observed in WT myocytes, consistent with BAG3 having effects upstream and at the level of the receptor. Resting membrane potential and action potential amplitude were unaffected but APD50 and APD90 were prolonged in shBAG3 myocytes. Protein levels of Ca(2+) entry molecules and other important excitation-contraction proteins were unchanged in myocytes with lower BAG3. Our findings that BAG3 is localized at the sarcolemma and t-tubules while modulating myocyte contraction and action potential duration through specific interaction with the β1-adrenergic receptor and L-type Ca(2+) channel provide novel insight into the role of BAG3 in cardiomyopathies and increased arrhythmia risks in heart failure.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.