166 results on '"Richard N. Kitsis"'
Search Results
2. Inducing and measuring apoptotic cell death in mouse pancreatic β-cells and in isolated islets
- Author
-
Wendy M. McKimpson and Richard N. Kitsis
- Subjects
Cell Biology ,Cell isolation ,Cell-based Assays ,Metabolism ,Science (General) ,Q1-390 - Abstract
Summary: Type 2 diabetes is mediated by insulin resistance and pancreatic β-cell failure, the latter reflecting a combination of β-cell dysfunction, dedifferentiation, and apoptosis. Quantification of β-cell apoptosis in diabetes can be challenging both with respect to methodology and selection of clinically relevant inducers and readouts. This protocol describes approaches to measure cell death in immortalized β-cells, primary mouse islet preparations, and pancreatic tissue. The resulting information may be useful for mechanistic studies and assessment of the contribution of β-cell death to pathogenesis.For complete details on the use and execution of this protocol, please refer to McKimpson et al. (2021).
- Published
- 2022
- Full Text
- View/download PDF
3. Editorial: Mitochondrial Dysfunction and Cardiovascular Diseases
- Author
-
Junichi Sadoshima, Richard N. Kitsis, and Sebastiano Sciarretta
- Subjects
mitochondrial dysfunction ,mitophagy ,cardiovascular diseases ,mitochondrial dynamics ,mitochondrial ROS ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Published
- 2021
- Full Text
- View/download PDF
4. OncomiR miR-182-5p Enhances Radiosensitivity by Inhibiting the Radiation-Induced Antioxidant Effect through SESN2 in Head and Neck Cancer
- Author
-
Min-Ying Lin, Yu-Chan Chang, Shan-Ying Wang, Muh-Hwa Yang, Chih-Hsien Chang, Michael Hsiao, Richard N. Kitsis, and Yi-Jang Lee
- Subjects
miR-182/96/183 cluster ,miR-182-5p ,head and neck squamous cell carcinoma ,radioresistance ,antioxidant ,SESN2 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Radiotherapy is routinely used for the treatment of head and neck squamous cell carcinoma (HNSCC). However, the therapeutic efficacy is usually reduced by acquired radioresistance and locoregional recurrence. In this study, The Cancer Genome Atlas (TCGA) analysis showed that radiotherapy upregulated the miR-182/96/183 cluster and that miR-182 was the most significantly upregulated. Overexpression of miR-182-5p enhanced the radiosensitivity of HNSCC cells by increasing intracellular reactive oxygen species (ROS) levels, suggesting that expression of the miR-182 family is beneficial for radiotherapy. By intersecting the gene targeting results from three microRNA target prediction databases, we noticed that sestrin2 (SESN2), a molecule resistant to oxidative stress, was involved in 91 genes predicted in all three databases to be directly recognized by miR-182-5p. Knockdown of SESN2 enhanced radiation-induced ROS and cytotoxicity in HNSCC cells. In addition, the radiation-induced expression of SESN2 was repressed by overexpression of miR-182-5p. Reciprocal expression of the miR-182-5p and SESN2 genes was also analyzed in the TCGA database, and a high expression of miR-182-5p combined with a low expression of SESN2 was associated with a better survival rate in patients receiving radiotherapy. Taken together, the current data suggest that miR-182-5p may regulate radiation-induced antioxidant effects and mediate the efficacy of radiotherapy.
- Published
- 2021
- Full Text
- View/download PDF
5. Uncontrolled angiogenic precursor expansion causes coronary artery anomalies in mice lacking Pofut1
- Author
-
Yidong Wang, Bingruo Wu, Pengfei Lu, Donghong Zhang, Brian Wu, Shweta Varshney, Gonzalo del Monte-Nieto, Zhenwu Zhuang, Rabab Charafeddine, Adam H. Kramer, Nicolas E. Sibinga, Nikolaos G. Frangogiannis, Richard N. Kitsis, Ralf H. Adams, Kari Alitalo, David J. Sharp, Richard P. Harvey, Pamela Stanley, and Bin Zhou
- Subjects
Science - Abstract
Though coronary arteries are crucial for heart function, the mechanisms guiding their formation are largely unknown. Here, Wang et al. identify a unique, endocardially-derived angiogenic precursor cell population for coronary artery formation in mice and show that a DLL4/NOTCH1/VEGFA/VEGFR2 signaling axis is key for coronary artery development.
- Published
- 2017
- Full Text
- View/download PDF
6. ARC is essential for maintaining pancreatic islet structure and β-cell viability during type 2 diabetes
- Author
-
Wendy M. McKimpson, Min Zheng, Streamson C. Chua, Jeffrey E. Pessin, and Richard N. Kitsis
- Subjects
Medicine ,Science - Abstract
Abstract Pancreatic β-cell loss through apoptosis is an important disease mechanism in type 2 diabetes. Apoptosis Repressor with CARD (ARC) is a cell death inhibitor that antagonizes multiple death programs. We previously reported that ARC is abundant in pancreatic β-cells and modulates survival of these cells in vitro. Herein we assessed the importance of endogenous ARC in maintaining islet structure and function in vivo. While generalized loss of ARC did not result in detectable abnormalities, its absence in ob/ob mice, a model of type 2 diabetes, induced a striking pancreatic phenotype: marked β-cell death, loss of β-cell mass, derangements of islet architecture, and impaired glucose-stimulated insulin secretion in vivo. These abnormalities contributed to worsening of hyperglycemia and glucose-intolerance in these mice. Mechanistically, the absence of ARC increased levels of C/EBP homologous protein (CHOP) in wild type isolated islets stimulated with ER stress and in ob/ob isolated islets at baseline. Deletion of CHOP in ob/ob; ARC −/− mice led to reversal of β-cell death and abnormalities in islet architecture. These data indicate that suppression of CHOP by endogenous levels of ARC is critical for β-cell viability and maintenance of normal islet structure in this model of type 2 diabetes.
- Published
- 2017
- Full Text
- View/download PDF
7. Troponin Release Following Brief Myocardial Ischemia
- Author
-
Dulguun Amgalan, MS, Ryan Pekson, PhD, and Richard N. Kitsis, MD
- Subjects
Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Published
- 2017
- Full Text
- View/download PDF
8. A Rab5 endosomal pathway mediates Parkin-dependent mitochondrial clearance
- Author
-
Babette C. Hammerling, Rita H. Najor, Melissa Q. Cortez, Sarah E. Shires, Leonardo J. Leon, Eileen R. Gonzalez, Daniela Boassa, Sébastien Phan, Andrea Thor, Rebecca E. Jimenez, Hong Li, Richard N. Kitsis, Gerald W. Dorn II, Junichi Sadoshima, Mark H. Ellisman, and Åsa B. Gustafsson
- Subjects
Science - Abstract
Damaged mitochondria are normally cleared through canonical and alternative autophagy pathways. Here, the authors report that mitochondria can be cleared through an autophagy-independent endosomal-lysosomal pathway that depends on Parkin-dependent sequestration of mitochondria in Rab5-positive early endosomes.
- Published
- 2017
- Full Text
- View/download PDF
9. Supplemental methods from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
Supplemental methods
- Published
- 2023
10. Supplemental Figure. 4 from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
Effects of Akt3/-S472 knockout and Akt3/+S472 or Akt1 overexpression on apoptosis
- Published
- 2023
11. Supplemental Figure.1 from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
Next generation sequencing of genomic DNA isolated from Akt3/+S472 and Akt3/-S472 knockout cell lines
- Published
- 2023
12. Supplementary Figure legends from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
Supplementary Figure legends
- Published
- 2023
13. Supplemental Figure. 5 from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
Akt3/-S472 overexpression in MDA-MB-436 cells suppresses tumor growth by inducing Bim expression and tumor apoptosis
- Published
- 2023
14. Supplemental Figure. 2 from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
Next generation sequencing of genomic DNA isolated from Akt3/+S472 and Akt3/-S472 knockout 3475 tumors
- Published
- 2023
15. Supplemental Figure. 3 from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
CRISPR mediated knockout of Akt3/-S472 in MDA-MB-231 cells increases mammary tumorigenesis
- Published
- 2023
16. Data from An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Rachel B. Hazan, Richard N. Kitsis, Larry Norton, Susan Fineberg, Yihong Wang, Jeffrey Segall, Greg R. Phillips, Wendy M. McKimpson, Dulguun Amgalan, Olivier D. Loudig, Outhiriaradjou Benard, Huizhi Liang, Jiahong Yao, and Kimita Suyama
- Abstract
The Akt pathway is a well-known promoter of tumor malignancy. Akt3 is expressed as two alternatively spliced variants, one of which lacks the key regulatory serine 472 phosphorylation site. Whereas the function of full-length Akt3 isoform (Akt3/+S472) is well-characterized, that of Akt3/−S472 isoform remains unknown. Despite being expressed at a substantially lower level than Akt3/+S472 in triple-negative breast cancer cells, specific ablation of Akt3/−S472 enhanced, whereas overexpression, suppressed mammary tumor growth, consistent with a significant association with patient survival duration relative to Akt3/+S472. These effects were due to striking induction of apoptosis, which was mediated by Bim upregulation, leading to conformational activation of Bax and caspase-3 processing. Bim accumulation was caused by marked endocytosis of EGF receptors with concomitant ERK attenuation, which stabilizes BIM. These findings demonstrate an unexpected function of an endogenously expressed Akt isoform in promoting, as opposed to suppressing, apoptosis, underscoring that Akt isoforms may exert dissonant functions in malignancy.Significance: These results illuminate an unexpected function for an endogenously expressed Akt isoform in promoting apoptosis, underscoring the likelihood that different Akt isoforms exert distinct functions in human cancer. Cancer Res; 78(1); 103–14. ©2017 AACR.
- Published
- 2023
17. Supplementary Figure 1 from Apoptosis Inhibitor ARC Promotes Breast Tumorigenesis, Metastasis, and Chemoresistance
- Author
-
Richard N. Kitsis, Jeffrey W. Pollard, Jeffrey Segall, Neal Ferrick, Benjamin Benson, Daniel Bamira, Tatiana Smirnova, Sumanta Goswami, and Christina M. Medina-Ramirez
- Abstract
PDF file - 42K
- Published
- 2023
18. Data from Apoptosis Inhibitor ARC Promotes Breast Tumorigenesis, Metastasis, and Chemoresistance
- Author
-
Richard N. Kitsis, Jeffrey W. Pollard, Jeffrey Segall, Neal Ferrick, Benjamin Benson, Daniel Bamira, Tatiana Smirnova, Sumanta Goswami, and Christina M. Medina-Ramirez
- Abstract
Apoptosis repressor with caspase recruitment domain (ARC) inhibits both death receptor- and mitochondrial/ER-mediated pathways of apoptosis. Although expressed mainly in terminally differentiated cells, ARC is markedly upregulated in a variety of human cancers, where its potential contributions have not yet been defined. In this study, we provide evidence of multiple critical pathophysiologic functions for ARC in breast carcinogenesis. In the polyoma middle T-antigen (PyMT) transgenic mouse model of breast cancer, in which endogenous ARC is strongly upregulated, deletion of the ARC-encoding gene nol3 decreased primary tumor burden without affecting tumor onset or multiplicity. More notably, ARC deficiency also limited tumor cell invasion and the number of circulating cancer cells, markedly reducing the number of lung metastases. Conversely, ectopic overexpression of ARC in a PyMT-derived metastatic breast cancer cell line increased invasion in vitro and lung metastasis in vivo. We confirmed these results in a humanized orthotopic model based on MDA-MB-231-derived LM2 metastatic breast cancer cells, in which RNAi-mediated knockdown of ARC levels was shown to reduce tumor volume, local invasion, and lung metastases. Lastly, we found that endogenous levels of ARC conferred chemoresistance in primary tumors and invading cell populations. Our results establish that ARC promotes breast carcinogenesis by driving primary tumor growth, invasion, and metastasis as well as by promoting chemoresistance in invasive cells. Cancer Res; 71(24); 7705–15. ©2011 AACR.
- Published
- 2023
19. Cardiac Myosin Heavy Chain Reporter Mice to Study Heart Development and Disease
- Author
-
Pengfei Lu, Bingruo Wu, Xuhui Feng, Wei Cheng, Richard N. Kitsis, and Bin Zhou
- Subjects
Mice ,Myosin Heavy Chains ,Physiology ,Myocardium ,Organogenesis ,Animals ,Heart ,Cardiology and Cardiovascular Medicine - Published
- 2022
20. Loss of apoptosis repressor with caspase recruitment domain (ARC) worsens high fat diet-induced hyperglycemia in mice
- Author
-
Richard N. Kitsis, Christine R. Schmidt, Steven E. Kahn, Sakeneh Zraika, Andrew T. Templin, Meghan F. Hogan, Rebecca L. Hull, and Nathalie Esser
- Subjects
medicine.medical_specialty ,Programmed cell death ,Arc (protein) ,biology ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Adipose tissue ,medicine.disease ,Endocrinology ,Apoptosis ,Internal medicine ,Diabetes mellitus ,medicine ,biology.protein ,Glucose homeostasis ,Caspase - Abstract
Apoptosis repressor with caspase recruitment domain (ARC) is an endogenous inhibitor of cell death signaling that is expressed in insulin-producing β cells. ARC has been shown to reduce β-cell death in response to diabetogenic stimuli in vitro, but its role in maintaining glucose homeostasis in vivo has not been fully established. Here we examined whether loss of ARC in FVB background mice exacerbates high fat diet (HFD)-induced hyperglycemia in vivo over 24 weeks. Prior to commencing 24-week HFD, ARC−/− mice had lower body weight than wild type (WT) mice. This body weight difference was maintained until the end of the study and was associated with decreased epididymal and inguinal adipose tissue mass in ARC−/− mice. Non-fasting plasma glucose was not different between ARC−/− and WT mice prior to HFD feeding, and ARC−/− mice displayed a greater increase in plasma glucose over the first 4 weeks of HFD. Plasma glucose remained elevated in ARC−/− mice after 16 weeks of HFD feeding, at which time it had returned to baseline in WT mice. Following 24 weeks of HFD, non-fasting plasma glucose in ARC−/− mice returned to baseline and was not different from WT mice. At this final time point, no differences were observed between genotypes in plasma glucose or insulin under fasted conditions or following intravenous glucose administration. However, HFD-fed ARC−/− mice exhibited significantly decreased β-cell area compared to WT mice. Thus, ARC deficiency delays, but does not prevent, metabolic adaptation to HFD feeding in mice, worsening transient HFD-induced hyperglycemia.
- Published
- 2021
21. Txnip C247S mutation protects the heart against acute myocardial infarction
- Author
-
Nobuhiro Mukai, Richard N. Kitsis, Yoshinobu Nakayama, Kristen Yang, Parth Patwari, Jun Yoshioka, and Bing F. Wang
- Subjects
0301 basic medicine ,Programmed cell death ,Thioredoxin-Interacting Protein ,Myocardial Infarction ,Gene Expression ,Mice, Transgenic ,030204 cardiovascular system & hematology ,Mitochondrion ,Article ,Cell Line ,Electrocardiography ,Mice ,03 medical and health sciences ,Adenosine Triphosphate ,Thioredoxins ,0302 clinical medicine ,Animals ,Hypoxia ,Molecular Biology ,Alleles ,Glyceraldehyde 3-phosphate dehydrogenase ,Disease Resistance ,chemistry.chemical_classification ,Reactive oxygen species ,biology ,Chemistry ,Cell biology ,Disease Models, Animal ,Oxidative Stress ,Glucose ,030104 developmental biology ,Amino Acid Substitution ,Organ Specificity ,Mutation ,biology.protein ,Disease Susceptibility ,Hypoxia-Inducible Factor 1 ,Signal transduction ,Thioredoxin ,Carrier Proteins ,Reactive Oxygen Species ,Cardiology and Cardiovascular Medicine ,Oxidation-Reduction ,Ubiquitin Thiolesterase ,Biomarkers ,TXNIP - Abstract
Rationale Thioredoxin-interacting protein (Txnip) is a novel molecular target with translational potential in diverse human diseases. Txnip has several established cellular actions including binding to thioredoxin, a scavenger of reactive oxygen species (ROS). It has been long recognized from in vitro evidence that Txnip forms a disulfide bridge through cysteine 247 (C247) with reduced thioredoxin to inhibit the anti-oxidative properties of thioredoxin. However, the physiological significance of the Txnip-thioredoxin interaction remains largely undefined in vivo. Objective A single mutation of Txnip, C247S, abolishes the binding of Txnip with thioredoxin. Using a conditional and inducible approach with a mouse model of a mutant Txnip that does not bind thioredoxin, we tested whether the interaction of thioredoxin with Txnip is required for Txnip's pro-oxidative or cytotoxic effects in the heart. Methods and results Overexpression of Txnip C247S in cells resulted in a reduction in ROS, due to an inability to inhibit thioredoxin. Hypoxia (1% O2, 24 h)-induced killing effects of Txnip were decreased by lower levels of cellular ROS in Txnip C247S-expressing cells compared with wild-type Txnip-expressing cells. Then, myocardial ischemic injuries were assessed in the animal model. Cardiomyocyte-specific Txnip C247S knock-in mice had better survival with smaller infarct size following myocardial infarction (MI) compared to control animals. The absence of Txnip's inhibition of thioredoxin promoted mitochondrial anti-oxidative capacities in cardiomyocytes, thereby protecting the heart from oxidative damage induced by MI. Furthermore, an unbiased RNA sequencing screen identified that hypoxia-inducible factor 1 signaling pathway was involved in Txnip C247S-mediated cardioprotective mechanisms. Conclusion Txnip is a cysteine-containing redox protein that robustly regulates the thioredoxin system via a disulfide bond-switching mechanism in adult cardiomyocytes. Our results provide the direct in vivo evidence that regulation of redox state by Txnip is a crucial component for myocardial homeostasis under ischemic stress.
- Published
- 2021
22. Multiple Cell Death Programs Contribute to Myocardial Infarction
- Author
-
Richard N. Kitsis, Felix G. Liang, and Xiaotong F. Jia
- Subjects
medicine.medical_specialty ,Programmed cell death ,Physiology ,caspase ,Myocardial Infarction ,necrosis ,Internal medicine ,medicine ,Humans ,Myocardial infarction ,Original Research ,Cell Death ,business.industry ,pyroptosis ,Pyroptosis ,medicine.disease ,inflammation ,Apoptosis ,Heart failure ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Cardiology ,Cardiology and Cardiovascular Medicine ,business - Abstract
Supplemental Digital Content is available in the text., Rationale: Pyroptosis is a morphologically and mechanistically distinct form of cell death and is characterized by GSDMD (gasdermin D) or GSDME (gasdermin E)-mediated necrosis with excessive inflammatory factor release. Cardiomyocyte necrosis and inflammation play key roles in the pathophysiology of myocardial ischemia/reperfusion (I/R) injury. However, whether cardiomyocytes undergo pyroptosis and the underlying mechanism in myocardial I/R injury remain unclear. Objective: We aimed to investigate the role of pyroptosis in myocardial I/R injury. Methods and Results: In vivo and in vitro experiments were used to investigate pyroptosis of cardiomyocyte and the associated mechanisms during I/R injury. Wild-type, Myh6-Cre, and cardiomyocyte-specific GSDMD-deficient male mice were subjected to I/R. Human peripheral blood samples were collected from patients with acute ST-segment–elevation myocardial infarction or control patients at 0, 1, and 24 hours after percutaneous coronary intervention in our department. The serum levels of GSDMD were measured by ELISA. Hypoxia/reoxygenation induced cardiomyocyte pyroptosis and the release of mature IL (interleukin)-18 but not IL-1β, which mechanistically resulted from GSDMD cleavage by caspase-11 in cardiomyocytes. Furthermore, GSDMD gene deletion blocked hypoxia/reoxygenation-induced cardiomyocyte pyroptosis and IL-18 release. GSDMD and its pyroptosis-inducing N-terminal fragment were upregulated in myocardial tissues after I/R injury. Immunofluorescence analysis showed that GSDMD was mainly localized in cardiomyocytes. GSDMD deficiency in cardiomyocytes significantly reduced the I/R-induced myocardial infarct size. Moreover, increased GSDMD serum levels were detected in patients exhibiting I/R injury 1 hour after percutaneous coronary intervention for ST-segment–elevation myocardial infarction. Conclusions: Our results show that GSDMD-mediated cardiomyocyte pyroptosis is a key event during myocardial I/R injury and that the caspase-11/GSDMD pathway may be essential to this process. Additionally, GSDMD inhibition significantly reduces cardiomyocyte pyroptosis and I/R-induced myocardial injury. Graphic Abstract: A graphic abstract is available for this article.
- Published
- 2021
23. ATG16L1 autophagy pathway regulates BAX protein levels and programmed cell death
- Author
-
Daorong Feng, Jeffrey E. Pessin, Richard N. Kitsis, Dulguun Amgalan, and Fenfen Chen
- Subjects
0301 basic medicine ,Autophagosome ,Programmed cell death ,Autophagy-Related Proteins ,Apoptosis ,Biochemistry ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Adipocyte ,SNAP23 ,Autophagy ,Animals ,Qc-SNARE Proteins ,Molecular Biology ,ATG16L1 ,bcl-2-Associated X Protein ,Gene knockdown ,030102 biochemistry & molecular biology ,Cell Biology ,Qb-SNARE Proteins ,Cell biology ,030104 developmental biology ,chemistry ,NIH 3T3 Cells ,Protein Binding ,Subcellular Fractions - Abstract
Previously we reported that adipocyte SNAP23 (synaptosome-associated protein of 23 kDa) deficiency blocks the activation of macroautophagy, leading to an increased abundance of BAX, a pro-death Bcl-2 family member, and activation and adipocyte cell death both in vitro and in vivo. Here, we found that knockdown of SNAP23 inhibited the association of the autophagosome regulators ATG16L1 and ATG9 compartments by nutrient depletion and reduced the formation of ATG16L1 membrane puncta. ATG16L1 knockdown inhibited autophagy flux and increased BAX protein levels by suppressing BAX degradation. The elevation in BAX protein had no effect on BAX activation or cell death in the nutrient-replete state. However, following nutrient depletion, BAX was activated with a concomitant induction of cell death. Co-immunoprecipitation analyses demonstrated that SNAP23 and ATG16L1 proteins form a stable complex independent of nutrient condition, whereas in the nutrient-depleted state, BAX binds to SNAP23 to form a ternary BAX–SNAP23–ATG16L1 protein complex. Taken together, these data support a model in which SNAP23 plays a crucial function as a scaffold for ATG16L1 necessary for the suppression of BAX activation and induction of the intrinsic cell death program.
- Published
- 2020
24. OncomiR miR-182-5p Enhances Radiosensitivity by Inhibiting the Radiation-Induced Antioxidant Effect through SESN2 in Head and Neck Cancer
- Author
-
Yu Chan Chang, Min Ying Lin, Michael Hsiao, Muh Hwa Yang, Shan Ying Wang, Chih Hsien Chang, Yi Jang Lee, and Richard N. Kitsis
- Subjects
antioxidant ,Physiology ,medicine.medical_treatment ,Clinical Biochemistry ,miR-182-5p ,RM1-950 ,medicine.disease_cause ,head and neck squamous cell carcinoma ,Biochemistry ,Article ,Radioresistance ,microRNA ,Medicine ,Radiosensitivity ,miR-182/96/183 cluster ,SESN2 ,Molecular Biology ,Gene knockdown ,business.industry ,Cell Biology ,Oncomir ,medicine.disease ,Head and neck squamous-cell carcinoma ,Radiation therapy ,radioresistance ,Cancer research ,Therapeutics. Pharmacology ,business ,Oxidative stress - Abstract
Radiotherapy is routinely used for the treatment of head and neck squamous cell carcinoma (HNSCC). However, the therapeutic efficacy is usually reduced by acquired radioresistance and locoregional recurrence. In this study, The Cancer Genome Atlas (TCGA) analysis showed that radiotherapy upregulated the miR-182/96/183 cluster and that miR-182 was the most significantly upregulated. Overexpression of miR-182-5p enhanced the radiosensitivity of HNSCC cells by increasing intracellular reactive oxygen species (ROS) levels, suggesting that expression of the miR-182 family is beneficial for radiotherapy. By intersecting the gene targeting results from three microRNA target prediction databases, we noticed that sestrin2 (SESN2), a molecule resistant to oxidative stress, was involved in 91 genes predicted in all three databases to be directly recognized by miR-182-5p. Knockdown of SESN2 enhanced radiation-induced ROS and cytotoxicity in HNSCC cells. In addition, the radiation-induced expression of SESN2 was repressed by overexpression of miR-182-5p. Reciprocal expression of the miR-182-5p and SESN2 genes was also analyzed in the TCGA database, and a high expression of miR-182-5p combined with a low expression of SESN2 was associated with a better survival rate in patients receiving radiotherapy. Taken together, the current data suggest that miR-182-5p may regulate radiation-induced antioxidant effects and mediate the efficacy of radiotherapy.
- Published
- 2021
25. Small Molecule Allosteric Inhibitors of BAX
- Author
-
Thomas P. Garner, Sheng Li, Evripidis Gavathiotis, Dulguun Amgalan, Richard N. Kitsis, and Denis E. Reyna
- Subjects
Models, Molecular ,Programmed cell death ,Allosteric regulation ,Apoptosis ,Chromosomal translocation ,Gas Chromatography-Mass Spectrometry ,Permeability ,Article ,03 medical and health sciences ,Proto-Oncogene Proteins ,Humans ,Amino Acid Sequence ,Molecular Biology ,Peptide sequence ,030304 developmental biology ,bcl-2-Associated X Protein ,0303 health sciences ,Binding Sites ,Chemistry ,Effector ,030302 biochemistry & molecular biology ,Cell Biology ,Small molecule ,Peptide Fragments ,Mitochondria ,3. Good health ,Cell biology ,Cytosol ,Proto-Oncogene Proteins c-bcl-2 ,Mitochondrial translocation - Abstract
BAX is a critical effector of the mitochondrial cell death pathway in response to a diverse range of stimuli in physiological and disease contexts. Upon binding by BH3-only proteins, cytosolic BAX undergoes conformational activation and translocation, resulting in mitochondrial outer membrane permeabilization. Efforts to rationally target BAX and develop inhibitors have been elusive, despite the clear therapeutic potential of inhibiting BAX-mediated cell death in a host of diseases. Here, we describe a class of small molecule BAX inhibitors, termed BAIs, which bind directly to a previously unrecognized pocket and allosterically inhibit BAX activation. BAI-binding around the hydrophobic helix α5 using hydrophobic and hydrogen bonding interactions stabilizes key areas of the hydrophobic core. BAIs inhibit conformational events in BAX activation that prevent BAX mitochondrial translocation and oligomerization. Our data highlight a novel paradigm for effective and selective pharmacological targeting of BAX to enable rational development of inhibitors of BAX-mediated cell death.
- Published
- 2019
26. Immune checkpoint inhibitor–associated myocarditis: manifestations and mechanisms
- Author
-
Andrew H. Lichtman, Richard N. Kitsis, Javid Moslehi, Lorenzo Galluzzi, and Arlene H. Sharpe
- Subjects
0301 basic medicine ,Myocarditis ,business.industry ,Immune checkpoint inhibitors ,Fulminant ,Inflammation ,General Medicine ,Review ,medicine.disease ,Cardiac cell ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Antigen ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Humans ,medicine.symptom ,business ,Immune Checkpoint Inhibitors ,Biomarkers - Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment of various cancers, including malignancies once considered untreatable. These agents, however, are associated with inflammation and tissue damage in multiple organs. Myocarditis has emerged as a serious ICI-associated toxicity, because, while seemingly infrequent, it is often fulminant and lethal. The underlying basis of ICI-associated myocarditis is not completely understood. While the importance of T cells is clear, the inciting antigens, why they are recognized, and the mechanisms leading to cardiac cell injury remain poorly characterized. These issues underscore the need for basic and clinical studies to define pathogenesis, identify predictive biomarkers, improve diagnostic strategies, and develop effective treatments. An improved understanding of ICI-associated myocarditis will provide insights into the equilibrium between the immune and cardiovascular systems.
- Published
- 2021
27. Exercise triggers CAPN1-mediated AIF truncation, inducing myocyte cell death in arrhythmogenic cardiomyopathy
- Author
-
Menotti Ruvo, Brittney Murray, Hugh Calkins, Carlos Bueno Beti, Matthew Miyamoto, Djahida Bedja, Cynthia A. James, Jacopo Agrimi, Gizem Keceli, Edon Melloni, Andrea Carpi, Richard N. Kitsis, Chulan Kwon, Brian O'Rourke, Crystal Tichnell, Fabio Di Lisa, Stephen P. Chelko, Nazareno Paolocci, Peter Andersen, Nunzianna Doti, Daniel P. Judge, An-Chi Wei, Marc K. Halushka, Angeliki Asimaki, Jeffrey E. Saffitz, and Nuria Amat-Codina
- Subjects
0301 basic medicine ,Programmed cell death ,congenital, hereditary, and neonatal diseases and abnormalities ,Necrosis ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Mitochondrion ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Myocyte ,Humans ,Cell Death ,Mitochondria ,Myocytes, Cardiac ,Apoptosis Inducing Factor ,Calpain ,Cardiomyopathies ,Physical Conditioning, Animal ,cardiovascular diseases ,Calpastatin ,Myocytes ,Muscle Cells ,Animal ,Chemistry ,General Medicine ,medicine.disease ,Physical Conditioning ,Cell biology ,Protein Transport ,030104 developmental biology ,Death, Sudden, Cardiac ,Apoptosis ,DNA fragmentation ,medicine.symptom ,biological phenomena, cell phenomena, and immunity ,Cardiac - Abstract
Myocyte death occurs in many inherited and acquired cardiomyopathies, including arrhythmogenic cardiomyopathy (ACM), a genetic heart disease plagued by the prevalence of sudden cardiac death. Individuals with ACM and harboring pathogenic desmosomal variants, such as desmoglein-2 (DSG2), often show myocyte necrosis with progression to exercise-associated heart failure. Here, we showed that homozygous Dsg2 mutant mice (Dsg2(mut/mut)), a model of ACM, die prematurely during swimming and display myocardial dysfunction and necrosis. We detected calcium (Ca(2+)) overload in Dsg2(mut/mut) hearts, which induced calpain-1 (CAPN1) activation, association of CAPN1 with mitochondria, and CAPN1-induced cleavage of mitochondrial-bound apoptosis-inducing factor (AIF). Cleaved AIF translocated to the myocyte nucleus triggering large-scale DNA fragmentation and cell death, an effect potentiated by mitochondrial-driven AIF oxidation. Posttranslational oxidation of AIF cysteine residues was due, in part, to a depleted mitochondrial thioredoxin-2 redox system. Hearts from exercised Dsg2(mut/mut) mice were depleted of calpastatin (CAST), an endogenous CAPN1 inhibitor, and overexpressing CAST in myocytes protected against Ca(2+) overload–induced necrosis. When cardiomyocytes differentiated from Dsg2(mut/mut) embryonic stem cells (ES-CMs) were challenged with β-adrenergic stimulation, CAPN1 inhibition attenuated CAPN1-induced AIF truncation. In addition, pretreatment of Dsg2(mut/mut) ES-CMs with an AIF-mimetic peptide, mirroring the cyclophilin-A (PPIA) binding site of AIF, blocked PPIA-mediated AIF-nuclear translocation, and reduced both apoptosis and necrosis. Thus, preventing CAPN1-induced AIF-truncation or barring binding of AIF to the nuclear chaperone, PPIA, may avert myocyte death and, ultimately, disease progression to heart failure in ACM and likely other forms of cardiomyopathies.
- Published
- 2021
28. Editorial: Mitochondrial Dysfunction and Cardiovascular Diseases
- Author
-
Junichi Sadoshima, Richard N. Kitsis, and Sebastiano Sciarretta
- Subjects
Mitochondrial ROS ,lcsh:Diseases of the circulatory (Cardiovascular) system ,Biology ,Cardiovascular Medicine ,mitochondrial ROS ,mitochondrial dynamics ,cardiovascular diseases ,Editorial ,mitophagy ,lcsh:RC666-701 ,Mitophagy ,mitochondrial dysfunction ,Cancer research ,Cardiology and Cardiovascular Medicine - Published
- 2020
29. Up‐regulation of cofilin‐1 in cell senescence associates with morphological change and p27 kip1 ‐mediated growth delay
- Author
-
Cheng-Han Tsai, Jhih Shian Lee, Pei Han Tai, Liang Tin Lin, Tzong-Jen Sheu, Jonathan D. Holz, Chun Yuan Chang, Wen Chien Huang, Meng Hsiu Wu, Yu Lou Wu, Yi Jang Lee, Richard N. Kitsis, and Bing Ze Lin
- Subjects
0301 basic medicine ,Senescence ,Aging ,Gene knockdown ,Reporter gene ,Cell ,macromolecular substances ,Cell Biology ,Biology ,environment and public health ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cofilin 1 ,medicine ,Enhancer ,TEAD1 ,Transcription factor ,030217 neurology & neurosurgery - Abstract
Morphological change is an explicit characteristic of cell senescence, but the underlying mechanisms remains to be addressed. Here, we demonstrated, after a survey of various actin-binding proteins, that the post-translational up-regulation of cofilin-1 was essential for the reduced rate of actin depolymerization morphological enlargement in senescent cells. Additionally, up-regulated cofilin-1 mainly existed in the serine-3 phosphorylated form, according to the 2D gel immunoblotting assay. The up-regulation of cofilin-1 was also detected in aged mammalian tissues. The over-expression of wild-type cofilin-1 and constitutively phosphorylated cofilin-1 promoted cell senescence with an increased cell size. Additionally, senescent phenotypes were also reduced by knockdown of total cofilin-1, which led to a decrease in phosphorylated cofilin-1. The senescence induced by the over-expression of cofilin-1 was dependent on p27Kip1 , but not on the p53 and p16INK4 expressions. The knockdown of p27Kip1 alleviated cell senescence induced by oxidative stress or replicative stress. We also found that the over-expression of cofilin-1 induced the expression of p27Kip1 through transcriptional suppression of the transcriptional enhancer factors domain 1 (TEAD1) transcription factor. The TEAD1 transcription factor played a transrepressive role in the p27Kip1 gene promoter, as determined by the promoter deletion reporter gene assay. Interestingly, the down-regulation of TEAD1 was accompanied by the up-regulation of cofilin-1 in senescence. The knockdown and restoration of TEAD1 in young cells and old cells could induce and inhibit p27Kip1 and senescent phenotypes, respectively. Taken together, the current data suggest that cofilin-1/TEAD1/p27Kip1 signaling is involved in senescence-related morphological change and growth arrest.
- Published
- 2020
30. 2042-P: Loss of Apoptosis Repressor with Caspase Recruitment Domain Promotes High-Fat Diet Induced Hyperglycemia In Vivo
- Author
-
Nathalie Esser, Christine R. Schmidt, Sakeneh Zraika, Meghan F. Hogan, Steven E. Kahn, Andrew T. Templin, Richard N. Kitsis, and Rebecca L. Hull
- Subjects
geography ,medicine.medical_specialty ,geography.geographical_feature_category ,Arc (protein) ,biology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Wild type ,Repressor ,Inhibitor of apoptosis ,Islet ,Endocrinology ,In vivo ,Apoptosis ,Internal medicine ,Internal Medicine ,biology.protein ,medicine ,business ,Caspase - Abstract
Apoptosis repressor with caspase recruitment domain (ARC) is an endogenous inhibitor of apoptosis signaling. We previously showed ARC is expressed in β cells and ameliorates amyloid-induced β-cell death in vitro. More recently, we found that wild type (WT) and whole-body ARC knockout (KO) mice on an FVB background fed a normal chow diet exhibit no differences in islet area, β-cell area, or α-cell area at 16 (n=5) or 35 weeks (n=7-8) of age. In the present study, we sought to determine whether ARC plays a role in maintaining glycemia in response to the challenge of high fat diet (HFD, 60% kcal from fat) feeding in vivo. At 12 weeks of age, just prior to starting 24 weeks of HFD, ARC KO mice displayed lower body weight than WT mice (0 wks: 26.2±0.6 vs. 30.6±0.8 g; p Disclosure A.T. Templin: None. C.R. Schmidt: None. M.F. Hogan: None. N. Esser: None. R.N. Kitsis: None. R.L. Hull: None. S. Zraika: Research Support; Self; Novartis Pharmaceuticals Corporation. S.E. Kahn: Advisory Panel; Self; Boehringer Ingelheim International GmbH, Eli Lilly and Company, Intarcia Therapeutics, Janssen Scientific Affairs, LLC., Merck & Co., Inc., Novo Nordisk A/S, Pfizer Inc. Funding U.S. Department of Veterans Affairs (I01-BX001060, IK2-BX004659); National Institutes of Health (P30DK017047)
- Published
- 2020
31. Conversion of the death inhibitor ARC to a killer activates pancreatic β cell death in diabetes
- Author
-
Richard N. Kitsis, Yun Chen, Jeremy Weinberger, David A. Lomas, Min Zheng, Wilson Lek Wen Tan, Wendy M. McKimpson, Jeffrey E. Pessin, Zenia Tiang, Alistair M. Jagger, Roger Foo, Streamson C. Chua, and James A. Irving
- Subjects
Male ,Programmed cell death ,Cytoplasm ,Cell ,Muscle Proteins ,Apoptosis ,Nerve Tissue Proteins ,Biology ,Serpin ,General Biochemistry, Genetics and Molecular Biology ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Insulin-Secreting Cells ,medicine ,Animals ,Humans ,Viability assay ,Molecular Biology ,030304 developmental biology ,Cell Nucleus ,Mice, Knockout ,0303 health sciences ,Arc (protein) ,Cell Biology ,Mice, Inbred C57BL ,Cytoskeletal Proteins ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,alpha 1-Antitrypsin ,Cancer research ,Female ,Apoptosis Regulatory Proteins ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Loss of insulin-secreting pancreatic β cells through apoptosis contributes to the progression of type 2 diabetes, but underlying mechanisms remain elusive. Here, we identify a pathway in which the cell death inhibitor ARC paradoxically becomes a killer during diabetes. While cytoplasmic ARC maintains β cell viability and pancreatic architecture, a pool of ARC relocates to the nucleus to induce β cell apoptosis in humans with diabetes and several pathophysiologically distinct mouse models. β cell death results through the coordinate downregulation of serpins (serine protease inhibitors) not previously known to be synthesized and secreted by β cells. Loss of the serpin α1-antitrypsin from the extracellular space unleashes elastase, triggering the disruption of β cell anchorage and subsequent cell death. Administration of α1-antitrypsin to mice with diabetes prevents β cell death and metabolic abnormalities. These data uncover a pathway for β cell loss in type 2 diabetes and identify an FDA-approved drug that may impede progression of this syndrome.
- Published
- 2020
32. Common mechanistic pathways in cancer and heart failure. A scientific roadmap on behalf of the Translational Research Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC)
- Author
-
Edoardo Bertero, Linda W. van Laake, Petar M. Seferovic, Ewa A. Jankowska, Javid Moslehi, Frank Ruschitzka, Richard N. Kitsis, Johann Bauersachs, Jean-Sébastien Hulot, Ovidiu Chioncel, Patrycja Nowak-Sliwinska, Joseph Pierre Aboumsallem, Dirk Jäger, Rudolf A. de Boer, Peter van der Meer, Douglas B. Sawyer, Dimitrios Farmakis, Lorenz H. Lehmann, Johannes Backs, Christoph Maack, Carlo G. Tocchetti, Suma H Konety, Massimo F Piepoli, Thomas Thum, Radek Pudil, Oliver J. Müller, Daniel J. Lenihan, James Larkin, Alexander R. Lyon, Pierre Dodion, Thomas M. Suter, Pietro Ameri, Thomas Eschenhagen, Antoni Bayes-Genis, Jelena Čelutkienė, Stephan von Haehling, Peter P. Rainer, Andrew J.S. Coats, Piotr Ponikowski, Stefan D. Anker, Stephane Heymans, Cardiovascular Centre (CVC), Restoring Organ Function by Means of Regenerative Medicine (REGENERATE), University Medical Center Groningen [Groningen] (UMCG), CIC - HEGP (CIC 1418), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPC), Paris-Centre de Recherche Cardiovasculaire (PARCC (UMR_S 970/ U970)), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPC), CArdiovasculaire Rénal Transplantation nEurovasculaire [Paris] (DMU CARTE), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), University of Naples Federico II, Universita degli studi di Genova, IRCCS Ospedale Policlinico San Martino [Genoa, Italy], Charité - UniversitätsMedizin = Charité - University Hospital [Berlin], Hannover Medical School [Hannover] (MHH), University Hospital of Würzburg, San Raffaele Pisana Scientific Institute for Resaearch, Hospitalisation, and Health Care, San Raffaele Institute Pisana, Vilnius University [Vilnius], University of Medicine and Pharmacy 'Carol Davila' Bucharest (UMPCD), Innate Pharma, Universitaetsklinikum Hamburg-Eppendorf = University Medical Center Hamburg-Eppendorf [Hamburg] (UKE), German Center for Cardiovascular Research (DZHK), Berlin Institute of Health (BIH), University of Cyprus (UCY), National and Kapodistrian University of Athens (NKUA), Hospital Universitari Germans Trias I Pujol [Badalona], Universitat Autònoma de Barcelona (UAB), Instituto de Salud Carlos III [Madrid] (ISC), Heidelberg University Hospital [Heidelberg], Wroclaw Medical University [Wrocław, Pologne], Albert Einstein College of Medicine [New York], University of Minnesota Medical School, University of Minnesota System, Royal Marsden NHS Foundation Trust, Universität Heidelberg [Heidelberg], Washington University in Saint Louis (WUSTL), Vanderbilt University Medical Center [Nashville], Vanderbilt University [Nashville], Kiel University, University of Geneva [Switzerland], University of Parma = Università degli studi di Parma [Parme, Italie], University Hospital Hradec Kralove, Medical University of Graz, University hospital of Zurich [Zurich], Maine Medical Center Research Institute (MMCRI), University of Belgrade [Belgrade], University of Bern, Utrecht University [Utrecht], University of Göttingen - Georg-August-Universität Göttingen, Maastricht University [Maastricht], Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven), Imperial College London, Cardiologie, MUMC+: MA Med Staf Spec Cardiologie (9), and RS: Carim - H02 Cardiomyopathy
- Subjects
Cardiac & Cardiovascular Systems ,SYMPATHETIC-NERVOUS-SYSTEM ,[SDV]Life Sciences [q-bio] ,angiogenesis ,cancer ,cardio-oncology ,cardiotoxicity ,clonal haematopoiesis ,extracellular matrix ,heart failure ,inflammation ,metabolism ,Disease ,Comorbidity ,030204 cardiovascular system & hematology ,Bioinformatics ,DISEASE ,0302 clinical medicine ,Risk Factors ,Neoplasms ,CACHEXIA ,INCREASED RISK ,Cancer ,ddc:615 ,TUMOR-GROWTH ,CLONAL HEMATOPOIESIS ,Extracellular matrix ,3. Good health ,Cardio‐oncology ,Cardio-oncology ,oncology ,Position Paper ,Cardiology and Cardiovascular Medicine ,Life Sciences & Biomedicine ,CARDIAC DYSFUNCTION ,DOXORUBICIN ,Translational research ,Context (language use) ,Heart failure ,Clonal haematopoiesis ,RADIATION-EXPOSURE ,ANTHRACYCLINE CARDIOTOXICITY ,03 medical and health sciences ,Genetic predisposition ,medicine ,Humans ,Clinical significance ,Inflammation ,Science & Technology ,business.industry ,Cardio‐ ,medicine.disease ,Cardiotoxicity ,Metabolism ,Cardiovascular System & Cardiology ,Personalized medicine ,Angiogenesis ,business - Abstract
The co‐occurrence of cancer and heart failure (HF) represents a significant clinical drawback as each disease interferes with the treatment of the other. In addition to shared risk factors, a growing body of experimental and clinical evidence reveals numerous commonalities in the biology underlying both pathologies. Inflammation emerges as a common hallmark for both diseases as it contributes to the initiation and progression of both HF and cancer. Under stress, malignant and cardiac cells change their metabolic preferences to survive, which makes these metabolic derangements a great basis to develop intersection strategies and therapies to combat both diseases. Furthermore, genetic predisposition and clonal haematopoiesis are common drivers for both conditions and they hold great clinical relevance in the context of personalized medicine. Additionally, altered angiogenesis is a common hallmark for failing hearts and tumours and represents a promising substrate to target in both diseases. Cardiac cells and malignant cells interact with their surrounding environment called stroma. This interaction mediates the progression of the two pathologies and understanding the structure and function of each stromal component may pave the way for innovative therapeutic strategies and improved outcomes in patients. The interdisciplinary collaboration between cardiologists and oncologists is essential to establish unified guidelines. To this aim, pre‐clinical models that mimic the human situation, where both pathologies coexist, are needed to understand all the aspects of the bidirectional relationship between cancer and HF. Finally, adequately powered clinical studies, including patients from all ages, and men and women, with proper adjudication of both cancer and cardiovascular endpoints, are essential to accurately study these two pathologies at the same time., We describe the co‐occurrence of cancer and heart failure (HF), their potential shared risk factors, and their pathophysiological mechanisms. We advocate intense interaction between cardiologists and oncologists to achieve unifying hypotheses and collaborative pre‐clinical and clinical studies.
- Published
- 2020
33. A small-molecule allosteric inhibitor of BAX protects against doxorubicin-induced cardiomyopathy
- Author
-
Jaehoon Lee, Victoria Margulets, Mounica Yanamandala, Evripidis Gavathiotis, Richard N. Kitsis, Victor Paulino, Swathi-Rao Narayanagari, Dulguun Amgalan, Yun Chen, Kelly Mitchell, Xiaotong F. Jia, Marco Scarlata, Luis Rivera Sanchez, Thomas P. Garner, Ulrich Steidl, Felix G. Liang, Rachel B. Hazan, John S. Condeelis, George S. Karagiannis, J. Jose Corbalan, Maja H. Oktay, Randall T. Peterson, Ryan Pekson, Lorrie A. Kirshenbaum, Aarti Asnani, Huizhi Liang, Andrea Lopez, Gaetano Santulli, Amgalan, Dulguun, Garner, Thomas P., Pekson, Ryan, Jia, Xiaotong F., Yanamandala, Mounica, Paulino, Victor, Liang, Felix G., Corbalan, J. Jose, Lee, Jaehoon, Chen, Yun, Karagiannis, George S., Sanchez, Luis Rivera, Liang, Huizhi, Narayanagari, Swathi-Rao, Mitchell, Kelly, Lopez, Andrea, Margulets, Victoria, Scarlata, Marco, Santulli, Gaetano, Asnani, Aarti, Peterson, Randall T., Hazan, Rachel B., Condeelis, John S., Oktay, Maja H., Steidl, Ulrich, Kirshenbaum, Lorrie A., Gavathiotis, Evripidi, and Kitsis, Richard N.
- Subjects
Cardioprotection ,Cancer Research ,Programmed cell death ,Necrosis ,Chemistry ,Cardiomyopathy ,Apoptosis ,Mitochondrion ,medicine.disease ,Article ,Mice ,Oncology ,Doxorubicin ,Cancer cell ,medicine ,Cancer research ,Animals ,medicine.symptom ,Cardiomyopathies ,Zebrafish ,medicine.drug ,bcl-2-Associated X Protein - Abstract
Doxorubicin remains an essential component of many cancer regimens, but its use is limited by lethal cardiomyopathy, which has been difficult to target, owing to pleiotropic mechanisms leading to apoptotic and necrotic cardiac cell death. Here we show that BAX is rate-limiting in doxorubicin-induced cardiomyopathy and identify a small-molecule BAX inhibitor that blocks both apoptosis and necrosis to prevent this syndrome. By allosterically inhibiting BAX conformational activation, this compound blocks BAX translocation to mitochondria, thereby abrogating both forms of cell death. When co-administered with doxorubicin, this BAX inhibitor prevents cardiomyopathy in zebrafish and mice. Notably, cardioprotection does not compromise the efficacy of doxorubicin in reducing leukemia or breast cancer burden in vivo, primarily due to increased priming of mitochondrial death mechanisms and higher BAX levels in cancer cells. This study identifies BAX as an actionable target for doxorubicin-induced cardiomyopathy and provides a prototype small-molecule therapeutic.
- Published
- 2020
34. Ryanodine Receptor Calcium Leak in Circulating B-Lymphocytes as a Biomarker in Heart Failure
- Author
-
Ellie J. Coromilas, Danielle L. Brunjes, Sarah J. Godfrey, Alexander Kushnir, Seth I. Sokol, Melana Yuzefpolskaya, Gaetano Santulli, Andrew R. Marks, Steven Reiken, Richard N. Kitsis, and Paolo C. Colombo
- Subjects
Male ,0301 basic medicine ,Leak ,Thiazepines ,chemistry.chemical_element ,030204 cardiovascular system & hematology ,Calcium ,Endoplasmic Reticulum ,Ventricular Function, Left ,Article ,Norepinephrine ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Animals ,Humans ,Medicine ,Calcium Signaling ,Systole ,Aged ,Heart Failure ,B-Lymphocytes ,business.industry ,Ryanodine receptor ,Disease progression ,Ryanodine Receptor Calcium Release Channel ,Middle Aged ,musculoskeletal system ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Case-Control Studies ,Heart failure ,cardiovascular system ,Cancer research ,Biomarker (medicine) ,Female ,Heart-Assist Devices ,Cardiology and Cardiovascular Medicine ,business ,tissues ,Intracellular - Abstract
Background: Advances in congestive heart failure (CHF) management depend on biomarkers for monitoring disease progression and therapeutic response. During systole, intracellular Ca 2+ is released from the sarcoplasmic reticulum into the cytoplasm through type-2 ryanodine receptor/Ca 2+ release channels. In CHF, chronically elevated circulating catecholamine levels cause pathological remodeling of type-2 ryanodine receptor/Ca 2+ release channels resulting in diastolic sarcoplasmic reticulum Ca 2+ leak and decreased myocardial contractility. Similarly, skeletal muscle contraction requires sarcoplasmic reticulum Ca 2+ release through type-1 ryanodine receptors (RyR1), and chronically elevated catecholamine levels in CHF cause RyR1-mediated sarcoplasmic reticulum Ca 2+ leak, contributing to myopathy and weakness. Circulating B-lymphocytes express RyR1 and catecholamine-responsive signaling cascades, making them a potential surrogate for defects in intracellular Ca 2+ handling because of leaky RyR channels in CHF. Methods: Whole blood was collected from patients with CHF, CHF following left-ventricular assist device implant, and controls. Blood was also collected from mice with ischemic CHF, ischemic CHF+S107 (a drug that specifically reduces RyR channel Ca 2+ leak), and wild-type controls. Channel macromolecular complex was assessed by immunostaining RyR1 immunoprecipitated from lymphocyte-enriched preparations. RyR1 Ca 2+ leak was assessed using flow cytometry to measure Ca 2+ fluorescence in B-lymphocytes in the absence and presence of RyR1 agonists that empty RyR1 Ca 2+ stores within the endoplasmic reticulum. Results: Circulating B-lymphocytes from humans and mice with CHF exhibited remodeled RyR1 and decreased endoplasmic reticulum Ca 2+ stores, consistent with chronic intracellular Ca 2+ leak. This Ca 2+ leak correlated with circulating catecholamine levels. The intracellular Ca 2+ leak was significantly reduced in mice treated with the Rycal S107. Patients with CHF treated with left-ventricular assist devices exhibited a heterogeneous response. Conclusions: In CHF, B-lymphocytes exhibit remodeled leaky RyR1 channels and decreased endoplasmic reticulum Ca 2+ stores consistent with chronic intracellular Ca 2+ leak. RyR1-mediated Ca 2+ leak in B-lymphocytes assessed using flow cytometry provides a surrogate measure of intracellular Ca 2+ handling and systemic sympathetic burden, presenting a novel biomarker for monitoring response to pharmacological and mechanical CHF therapy.
- Published
- 2018
35. An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis
- Author
-
Huizhi Liang, Greg R. Phillips, Olivier Loudig, Jiahong Yao, Yihong Wang, Richard N. Kitsis, Kimita Suyama, Outhiriaradjou Benard, Jeffrey E. Segall, Susan Fineberg, Wendy M. McKimpson, Dulguun Amgalan, Rachel B. Hazan, and Larry Norton
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Gene isoform ,Cancer Research ,Mice, Nude ,Apoptosis ,Triple Negative Breast Neoplasms ,Biology ,Article ,AKT3 ,03 medical and health sciences ,Downregulation and upregulation ,Cell Line, Tumor ,Serine ,Animals ,Humans ,Protein Isoforms ,Phosphorylation ,Protein kinase B ,PI3K/AKT/mTOR pathway ,bcl-2-Associated X Protein ,Mammary tumor ,Bcl-2-Like Protein 11 ,Caspase 3 ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,Cancer research ,Female ,RNA Splice Sites ,Proto-Oncogene Proteins c-akt - Abstract
The Akt pathway is a well-known promoter of tumor malignancy. Akt3 is expressed as two alternatively spliced variants, one of which lacks the key regulatory serine 472 phosphorylation site. Whereas the function of full-length Akt3 isoform (Akt3/+S472) is well-characterized, that of Akt3/−S472 isoform remains unknown. Despite being expressed at a substantially lower level than Akt3/+S472 in triple-negative breast cancer cells, specific ablation of Akt3/−S472 enhanced, whereas overexpression, suppressed mammary tumor growth, consistent with a significant association with patient survival duration relative to Akt3/+S472. These effects were due to striking induction of apoptosis, which was mediated by Bim upregulation, leading to conformational activation of Bax and caspase-3 processing. Bim accumulation was caused by marked endocytosis of EGF receptors with concomitant ERK attenuation, which stabilizes BIM. These findings demonstrate an unexpected function of an endogenously expressed Akt isoform in promoting, as opposed to suppressing, apoptosis, underscoring that Akt isoforms may exert dissonant functions in malignancy. Significance: These results illuminate an unexpected function for an endogenously expressed Akt isoform in promoting apoptosis, underscoring the likelihood that different Akt isoforms exert distinct functions in human cancer. Cancer Res; 78(1); 103–14. ©2017 AACR.
- Published
- 2018
36. Abstract 1334: CCR5 inhibitors enhance doxorubicin-induced breast cancer cell killing while reducing cardiotoxicity
- Author
-
Xuanmao Jiao, Joseph C. Wu, Zhiping Li, Richard G. Pestell, Sean Lal, Anthony W. Ashton, Richard N. Kitsis, and Cristobal G. dos Remedios
- Subjects
Cancer Research ,Cardiotoxicity ,Chemotherapy ,biology ,business.industry ,DNA repair ,Topoisomerase ,medicine.medical_treatment ,Cancer ,medicine.disease ,Transplantation ,Breast cancer ,Oncology ,biology.protein ,Cancer research ,Medicine ,Doxorubicin ,business ,medicine.drug - Abstract
Cardiac toxicity from chemotherapy, which contributes substantially to morbidity and mortality in cancer survivors is dose-dependent, but sensitivity can vary greatly between patients. Doxorubicin (DOX) reduces ventricular ejection fraction to 1.5 million women develop breast cancer annually, it is estimated that >50,000 women/year will develop severe cardiotoxicity. With cancer survivors estimated at 19 million in the USA by 2025, DOX-induced cardiotoxicity is considered part of the “cardio-oncology epidemic”. Two cellular activities: DNA damage, causing double-strand breaks (DSBs) following poisoning of topoisomerase II (Topo II), and chromatin damage, mediated through histone eviction, are required for cardiotoxicity. The only FDA approved preventive, dexrazoxane reduces the anti-cancer efficacy of the DOX-based chemotherapeutic agents has the concerning side effects of myelotoxicity and a possible increased prevalence of secondary malignancies. We previously showed that the G-protein coupled receptor CCR5 is expressed on both immune and epithelial cells of ~50% of human breast cancers (BCa), thereby inducing cancer “stemness”, cell survival and DNA repair and a pro-metastatic phenotype. Moreover, CCR5 expression is enhanced in DOX-resistant BCa cells and CCR5 inhibitors (CCR5i) enhance DOX-induced cell death of BCa cells. Whilst expression of CCR5 and its ligands (CCL3 and CCL5) were induced by DOX-treatment in cardiac myocytes in both a murine model of DOX-cardiac toxicity and in the hearts of patients undergoing cardiac transplantation for DOX-induced cardiomyopathy. Unlike in BCa cells (where the enhanced CCR5 expression is transcriptionaly regulated) the increased expression in the myocardium occurs in the absence of altered mRNA levels suggesting epigenetic control over CCR5 expression during DOX-induced cardiotoxicity. In the myocardium CCR5 signaling promotes cardiac remodeling and heart failure, opposite that in BCa where CCR5 signaling promotes survival. Not surprisingly, CCR5i protected human iPSC-derived cardiomyocytes and isolated canine cardiomyocytes from DOX-induced cell death using nuclear fragmentation (hypodiploid peak) and mitochondrial function (MitoGlo reagent) as endpoints. Further, in a murine model of DOX cardiotoxicity (3 mg/kgx8 doses over 2 weeks) CCR5i substantially reduced (>90%) DOX-induced cardiac dysfunction and mortality in mice. CCR5 inhibitors (CCR5i) are “dual function” compounds that provide both cardiac protection and enhanced breast cancer cell killing in the presence of DNA damaging chemotherapy agents. Our studies may have a broad impact by identifying a novel approach to both enhancing therapeutic efficacy and providing cardioprotection from DNA damaging agents that are widely used in cancer treatment. Citation Format: Anthony W. Ashton, Xuanmao Jiao, Zhiping Li, Joseph C. Wu, Cristobal dos Remedios, Sean Lal, Richard Kitsis, Richard G. Pestell. CCR5 inhibitors enhance doxorubicin-induced breast cancer cell killing while reducing cardiotoxicity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1334.
- Published
- 2021
37. Overcoming the Roadblocks to Cardiac Cell Therapy Using Tissue Engineering
- Author
-
Nenad Bursac, Mounica Yanamandala, Timothy J. Kamp, Richard N. Kitsis, Roberto Bolli, Joshua M. Hare, Young Sup Yoon, Wuqiang Zhu, Ho-Wook Jun, Daniel J. Garry, Gerald W. Dorn, Jianyi Zhang, and Sumanth D. Prabhu
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Heart Diseases ,Tissue Engineering ,business.industry ,medicine.medical_treatment ,Cell- and Tissue-Based Therapy ,Stem-cell therapy ,medicine.disease ,Biocompatible material ,Article ,Cardiac cell ,03 medical and health sciences ,030104 developmental biology ,Tissue engineering ,Internal medicine ,Heart failure ,Infarcted heart ,medicine ,Cardiology ,Humans ,Myocardial infarction ,Stem cell ,Cardiology and Cardiovascular Medicine ,business - Abstract
Transplantations of various stem cells or their progeny have repeatedly improved cardiac performance in animal models of myocardial injury; however, the benefits observed in clinical trials have been generally less consistent. Some of the recognized challenges are poor engraftment of implanted cells and, in the case of human cardiomyocytes, functional immaturity and lack of electrical integration, leading to limited contribution to the heart’s contractile activity and increased arrhythmogenic risks. Advances in tissue and genetic engineering techniques are expected to improve the survival and integration of transplanted cells, and to support structural, functional, and bioenergetic recovery of the recipient hearts. Specifically, application of a prefabricated cardiac tissue patch to prevent dilation and to improve pumping efficiency of the infarcted heart offers a promising strategy for making stem cell therapy a clinical reality.
- Published
- 2017
38. Apoptosis Repressor With Caspase Recruitment Domain Ameliorates Amyloid-Induced β-Cell Apoptosis and JNK Pathway Activation
- Author
-
Rebecca L. Hull, Meghan F. Hogan, Steven E. Kahn, Sakeneh Zraika, Richard N. Kitsis, Michael T. Crow, Daniel T. Meier, Tanya Samarasekera, Andrew T. Templin, and Mahnaz Mellati
- Subjects
0301 basic medicine ,Male ,Amyloid ,Endocrinology, Diabetes and Metabolism ,Blotting, Western ,Repressor ,Muscle Proteins ,Apoptosis ,Mice, Transgenic ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Insulin-Secreting Cells ,Internal Medicine ,Animals ,Immunoprecipitation ,Caspase ,Cells, Cultured ,geography ,Arc (protein) ,geography.geographical_feature_category ,biology ,Kinase ,Reverse Transcriptase Polymerase Chain Reaction ,JNK Mitogen-Activated Protein Kinases ,Islet ,Molecular biology ,030104 developmental biology ,Islet Studies ,030220 oncology & carcinogenesis ,biology.protein ,Phosphorylation ,Female ,Apoptosis Regulatory Proteins - Abstract
Islet amyloid is present in more than 90% of individuals with type 2 diabetes, where it contributes to β-cell apoptosis and insufficient insulin secretion. Apoptosis repressor with caspase recruitment domain (ARC) binds and inactivates components of the intrinsic and extrinsic apoptosis pathways and was recently found to be expressed in islet β-cells. Using a human islet amyloid polypeptide transgenic mouse model of islet amyloidosis, we show ARC knockdown increases amyloid-induced β-cell apoptosis and loss, while ARC overexpression decreases amyloid-induced apoptosis, thus preserving β-cells. These effects occurred in the absence of changes in islet amyloid deposition, indicating ARC acts downstream of amyloid formation. Because islet amyloid increases c-Jun N-terminal kinase (JNK) pathway activation, we investigated whether ARC affects JNK signaling in amyloid-forming islets. We found ARC knockdown enhances JNK pathway activation, whereas ARC overexpression reduces JNK, c-Jun phosphorylation, and c-Jun target gene expression (Jun and Tnf). Immunoprecipitation of ARC from mouse islet lysates showed ARC binds JNK, suggesting interaction between JNK and ARC decreases amyloid-induced JNK phosphorylation and downstream signaling. These data indicate that ARC overexpression diminishes amyloid-induced JNK pathway activation and apoptosis in the β-cell, a strategy that may reduce β-cell loss in type 2 diabetes.
- Published
- 2017
39. Troponin Release Following Brief Myocardial Ischemia
- Author
-
Ryan Pekson, Richard N. Kitsis, and Dulguun Amgalan
- Subjects
0301 basic medicine ,lcsh:Diseases of the circulatory (Cardiovascular) system ,medicine.medical_specialty ,Programmed cell death ,Necrosis ,Myocardial ischemia ,biology ,business.industry ,Ischemia ,030204 cardiovascular system & hematology ,medicine.disease ,Troponin ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,lcsh:RC666-701 ,Apoptosis ,Internal medicine ,medicine ,Cardiology ,biology.protein ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Published
- 2017
40. A myeloid tumor suppressor role for NOL3
- Author
-
Kira Gritsman, Ulrich Steidl, Hideki Makishima, Britta Will, Richard T. Piszczatowski, Wendy M. McKimpson, Cassandra M. Hirsch, Kelly Mitchell, Christine McMahon, Boris Bartholdy, Swathi Rao Narayanagari, Richard N. Kitsis, Robert F. Stanley, Tihomira I. Todorova, Jaroslaw P. Maciejewski, and Dagmar Walter
- Subjects
0301 basic medicine ,Myeloid ,biology ,Immunology ,Cell ,CD34 ,medicine.disease ,Phenotype ,3. Good health ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,biology.protein ,Cancer research ,Immunology and Allergy ,Cyclin-dependent kinase 6 ,Myelofibrosis ,Janus kinase - Abstract
Despite the identification of several oncogenic driver mutations leading to constitutive JAK–STAT activation, the cellular and molecular biology of myeloproliferative neoplasms (MPN) remains incompletely understood. Recent discoveries have identified underlying disease-modifying molecular aberrations contributing to disease initiation and progression. Here, we report that deletion of Nol3 (Nucleolar protein 3) in mice leads to an MPN resembling primary myelofibrosis (PMF). Nol3−/− MPN mice harbor an expanded Thy1+LSK stem cell population exhibiting increased cell cycling and a myelomonocytic differentiation bias. Molecularly, this phenotype is mediated by Nol3−/−-induced JAK–STAT activation and downstream activation of cyclin-dependent kinase 6 (Cdk6) and Myc. Nol3−/− MPN Thy1+LSK cells share significant molecular similarities with primary CD34+ cells from PMF patients. NOL3 levels are decreased in CD34+ cells from PMF patients, and the NOL3 locus is deleted in a subset of patients with myeloid malignancies. Our results reveal a novel genetic PMF-like mouse model and identify a tumor suppressor role for NOL3 in the pathogenesis of myeloid malignancies.
- Published
- 2017
41. A Rab5 endosomal pathway mediates Parkin-dependent mitochondrial clearance
- Author
-
Daniela Boassa, Babette C. Hammerling, Richard N. Kitsis, Sebastien Phan, Hong Li, Rebecca E. Jimenez, Åsa B. Gustafsson, Sarah E. Shires, Mark H. Ellisman, Melissa Q. Cortez, Gerald W. Dorn, Eileen R. Gonzalez, Junichi Sadoshima, Andrea Thor, Rita H. Najor, and Leonardo J. Leon
- Subjects
Male ,0301 basic medicine ,General Physics and Astronomy ,Apoptosis ,Mitochondrion ,Parkin ,Autophagy-Related Protein 5 ,Rats, Sprague-Dawley ,Mice ,0302 clinical medicine ,Ubiquitin ,Myocyte ,Myocytes, Cardiac ,RNA, Small Interfering ,Mice, Knockout ,Multidisciplinary ,biology ,Chemistry ,Mitophagy ,Mitochondria ,Cell biology ,Gene Knockdown Techniques ,Beclin-1 ,Female ,Signal Transduction ,Programmed cell death ,Endosome ,Ubiquitin-Protein Ligases ,Science ,Primary Cell Culture ,Endosomes ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Organelle ,Autophagy ,Animals ,rab5 GTP-Binding Proteins ,Endosomal Sorting Complexes Required for Transport ,fungi ,Ubiquitination ,General Chemistry ,Fibroblasts ,Rats ,Mice, Inbred C57BL ,Microscopy, Electron ,030104 developmental biology ,biology.protein ,Lysosomes ,030217 neurology & neurosurgery - Abstract
Damaged mitochondria pose a lethal threat to cells that necessitates their prompt removal. The currently recognized mechanism for disposal of mitochondria is autophagy, where damaged organelles are marked for disposal via ubiquitylation by Parkin. Here we report a novel pathway for mitochondrial elimination, in which these organelles undergo Parkin-dependent sequestration into Rab5-positive early endosomes via the ESCRT machinery. Following maturation, these endosomes deliver mitochondria to lysosomes for degradation. Although this endosomal pathway is activated by stressors that also activate mitochondrial autophagy, endosomal-mediated mitochondrial clearance is initiated before autophagy. The autophagy protein Beclin1 regulates activation of Rab5 and endosomal-mediated degradation of mitochondria, suggesting cross-talk between these two pathways. Abrogation of Rab5 function and the endosomal pathway results in the accumulation of stressed mitochondria and increases susceptibility to cell death in embryonic fibroblasts and cardiac myocytes. These data reveal a new mechanism for mitochondrial quality control mediated by Rab5 and early endosomes., Damaged mitochondria are normally cleared through canonical and alternative autophagy pathways. Here, the authors report that mitochondria can be cleared through an autophagy-independent endosomal-lysosomal pathway that depends on Parkin-dependent sequestration of mitochondria in Rab5-positive early endosomes.
- Published
- 2017
42. RCAN1–Calcineurin Axis and the Set-Point for Myocardial Damage During Ischemia-Reperfusion
- Author
-
J. Jose Corbalan and Richard N. Kitsis
- Subjects
Dynamins ,Male ,0301 basic medicine ,Calmodulin ,Physiology ,Phosphatase ,Muscle Proteins ,Myocardial Reperfusion Injury ,Mitochondrial Dynamics ,Mitochondria, Heart ,Article ,Cell Line ,Rats, Sprague-Dawley ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Receptor ,Transcription factor ,Cells, Cultured ,Heart valve formation ,biology ,Calpain ,Chemistry ,Calcineurin ,Myocardium ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,NFAT ,medicine.disease ,Rats ,Mitochondria ,Cell biology ,Mice, Inbred C57BL ,Oxygen ,DNA-Binding Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Down Syndrome ,Cardiology and Cardiovascular Medicine ,Reperfusion injury - Abstract
RATIONALE: The Regulator of Calcineurin 1 (RCAN1) inhibits calcineurin (CN), a Ca(2+)-activated protein phosphatase important in cardiac remodeling. In humans, RCAN1 is located on chromosome 21 in proximity to the “Down syndrome critical region.” The hearts and brains of Rcan1 KO mice are more susceptible to damage from ischemia/reperfusion (I/R), however, the underlying cause is not known. OBJECTIVE: Mitochondria are key mediators of I/R damage. The goal of these studies was to determine the impact of RCAN1 on mitochondrial dynamics and function. METHODS AND RESULTS: Using both neonatal and isolated adult cardiomyocytes, we show that, when RCAN1 is depleted, the mitochondrial network is more fragmented due to increased CN-dependent activation of the fission protein, Dynamin-1-Like (DRP1). Mitochondria in RCAN1-depleted cardiomyocytes have reduced membrane potential, O(2) consumption, and generation of reactive oxygen species, as well as a reduced capacity for mitochondrial Ca(2+) uptake. RCAN1-depleted cardiomyocytes were more sensitive to I/R, however, pharmacological inhibition of CN, DRP1, or calpains (Ca(2+)-activated proteases) restored protection, suggesting that, in the absence of RCAN1, calpain-mediated damage following I/R is greater due to a decrease in the capacity of mitochondria to buffer cytoplasmic Ca(2+). Increasing RCAN1 levels by adenoviral infection was sufficient to enhance fusion and confer protection from I/R. To examine the impact of more modest, and biologically relevant, increases in RCAN1, we compared the mitochondrial network in induced pluripotent stem cells (iPSC) derived from individuals with Down syndrome to that of isogenic, disomic controls. Mitochondria were more fused and O(2) consumption was greater in the trisomic iPSC, however, coupling efficiency and metabolic flexibility was compromised compared to disomic. Depletion of RCAN1 from trisomic iPSC was sufficient to normalize mitochondrial dynamics and function. CONCLUSIONS: RCAN1 helps maintain a more interconnected mitochondrial network and maintaining appropriate RCAN1 levels is important to human health and disease.
- Published
- 2018
43. Unlocking the Secrets of Mitochondria in the Cardiovascular System: Path to a Cure in Heart Failure—A Report from the 2018 National Heart, Lung, and Blood Institute Workshop
- Author
-
Rong Tian, Wilson S. Colucci, Zoltan Arany, Markus M. Bachschmid, Scott W. Ballinger, Sihem Boudina, James E. Bruce, David W. Busija, Sergey Dikalov, Gerald W. Dorn, Zorina S. Galis, Roberta A. Gottlieb, Daniel P. Kelly, Richard N. Kitsis, Mark J. Kohr, Daniel Levy, E. Douglas Lewandowski, Joseph M. McClung, Daria Mochly-Rosen, Kevin D. O’Brien, Brian O’Rourke, Joon-Young Park, Peipei Ping, Michael N. Sack, Shey-Shing Sheu, Yang Shi, Sruti Shiva, Douglas C. Wallace, Robert G. Weiss, Hilary J. Vernon, Renee Wong, Lisa Schwartz Longacre, and Scarlet Shi
- Subjects
Research Report ,medicine.medical_specialty ,Biomedical Research ,030204 cardiovascular system & hematology ,Mitochondrion ,Cardiovascular System ,Article ,Education ,Pathogenesis ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,medicine ,Humans ,030304 developmental biology ,Heart Failure ,0303 health sciences ,Lung ,business.industry ,medicine.disease ,United States ,Mitochondria ,medicine.anatomical_structure ,Heart failure ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,National Heart, Lung, and Blood Institute (U.S.) - Abstract
Mitochondria have emerged as a central factor in the pathogenesis and progression of heart failure, and other cardiovascular diseases, as well, but no therapies are available to treat mitochondrial dysfunction. The National Heart, Lung, and Blood Institute convened a group of leading experts in heart failure, cardiovascular diseases, and mitochondria research in August 2018. These experts reviewed the current state of science and identified key gaps and opportunities in basic, translational, and clinical research focusing on the potential of mitochondria-based therapeutic strategies in heart failure. The workshop provided short- and long-term recommendations for moving the field toward clinical strategies for the prevention and treatment of heart failure and cardiovascular diseases by using mitochondria-based approaches.
- Published
- 2019
44. Fundamental Mechanisms of Regulated Cell Death and Implications for Heart Disease
- Author
-
Richard N. Kitsis, Andreas Linkermann, Dulguun Amgalan, Dominic P. Del Re, and Qinghang Liu
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,Programmed cell death ,Necrosis ,Heart Diseases ,Physiology ,Necroptosis ,Autophagy-Related Proteins ,Apoptosis ,Review ,Mitochondria, Heart ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,medicine ,Autophagy ,Pyroptosis ,Animals ,Humans ,Molecular Biology ,Heart metabolism ,Cell Death ,business.industry ,Myocardium ,General Medicine ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,medicine.symptom ,Signal transduction ,business ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
Twelve regulated cell death programs have been described. We review in detail the basic biology of nine including death receptor-mediated apoptosis, death receptor-mediated necrosis (necroptosis), mitochondrial-mediated apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death. This is followed by a dissection of the roles of these cell death programs in the major cardiac syndromes: myocardial infarction and heart failure. The most important conclusion relevant to heart disease is that regulated forms of cardiomyocyte death play important roles in both myocardial infarction with reperfusion (ischemia/reperfusion) and heart failure. While a role for apoptosis in ischemia/reperfusion cannot be excluded, regulated forms of necrosis, through both death receptor and mitochondrial pathways, are critical. Ferroptosis and parthanatos are also likely important in ischemia/reperfusion, although it is unclear if these entities are functioning as independent death programs or as amplification mechanisms for necrotic cell death. Pyroptosis may also contribute to ischemia/reperfusion injury, but potentially through effects in non-cardiomyocytes. Cardiomyocyte loss through apoptosis and necrosis is also an important component in the pathogenesis of heart failure and is mediated by both death receptor and mitochondrial signaling. Roles for immunogenic cell death in cardiac disease remain to be defined but merit study in this era of immune checkpoint cancer therapy. Biology-based approaches to inhibit cell death in the various cardiac syndromes are also discussed.
- Published
- 2019
45. Correcting mitochondrial fusion by manipulating mitofusin conformations
- Author
-
Guohua Gong, Daria Mochly-Rosen, Nir Qvit, Ann Benz, Richard N. Kitsis, Sara K. Donnelly, Gerald W. Dorn, Evripidis Gavathiotis, Antonietta Franco, Steven Mennerick, Opher S. Kornfeld, Yun Chen, Julie Fleischer, Louis Hodgson, and Nikolaos Biris
- Subjects
Models, Molecular ,0301 basic medicine ,Protein Conformation ,MFN2 ,Mitochondrion ,Biology ,Mitochondrial Dynamics ,Article ,Permeability ,GTP Phosphohydrolases ,Mice ,03 medical and health sciences ,Mitofusin-2 ,0302 clinical medicine ,Charcot-Marie-Tooth Disease ,Organelle ,Animals ,Gene ,Cells, Cultured ,Neurons ,Multidisciplinary ,Fibroblasts ,Mitochondria ,Cell biology ,030104 developmental biology ,mitochondrial fusion ,Biochemistry ,Mitochondrial fission ,Peptides ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Mitochondria are dynamic organelles that exchange contents and undergo remodelling during cyclic fusion and fission. Genetic mutations in MFN2 (the gene encoding mitofusin 2) interrupt mitochondrial fusion and cause the untreatable neurodegenerative condition Charcot-Marie-Tooth disease type 2A (CMT2A). It has not yet been possible to directly modulate mitochondrial fusion, in part because the structural basis of mitofusin function is not completely understood. Here we show that mitofusins adopt either a fusion-constrained or a fusion-permissive molecular conformation, directed by specific intramolecular binding interactions, and demonstrate that mitofusin-dependent mitochondrial fusion can be regulated in mouse cells by targeting these conformational transitions. On the basis of this model, we engineered a cell-permeant minipeptide to destabilize the fusion-constrained conformation of mitofusin and promote the fusion-permissive conformation, reversing mitochondrial abnormalities in cultured fibroblasts and neurons that harbour CMT2A-associated genetic defects. The relationship between the conformational plasticity of mitofusin 2 and mitochondrial dynamism reveals a central mechanism that regulates mitochondrial fusion, the manipulation of which can correct mitochondrial pathology triggered by defective or imbalanced mitochondrial dynamics.
- Published
- 2016
46. Grounding Cardio-Oncology in Basic and Clinical Science
- Author
-
Javid Moslehi, Dulguun Amgalan, and Richard N. Kitsis
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Pathology ,Biomedical Research ,Myocarditis ,Population ,Disease ,030204 cardiovascular system & hematology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pharmacotherapy ,Cancer Survivors ,Neoplasms ,Physiology (medical) ,medicine ,Humans ,Intensive care medicine ,education ,Cause of death ,education.field_of_study ,business.industry ,Vascular disease ,Cancer ,medicine.disease ,030104 developmental biology ,Cardiovascular Diseases ,Personalized medicine ,Cardiology and Cardiovascular Medicine ,business - Abstract
The development of cardio-oncology as a clinical field has been driven by improved therapeutics resulting in a dramatic increase in the number of cancer survivors (≈15 500 000 in the United States alone) many of whom experience cardiovascular complications related to their cancer treatments.1 Both traditional and targeted therapies can affect the cardiovascular system, resulting in hypertension, heart failure, myocarditis, arrhythmias, vascular disease, and thrombosis. Although recurrence of cancer is the eventual cause of death in many cancer survivors, cardiovascular disease is responsible for substantial morbidity and mortality in this population. Thus, effective means of mitigating cardiovascular complications of cancer therapies would have substantial impact on health. Unfortunately, definitive strategies to predict, prevent, monitor, and treat chemotherapy-induced cardiovascular syndromes are currently lacking for several reasons. First, mechanistically distinct cancer therapies can cause heterogeneous cardiovascular sequelae. Second, molecular mechanisms that mediate these syndromes are poorly understood. Last, evidence-based knowledge pertaining to some of the most important clinical questions is not yet available. Because of this situation, most clinical guidelines are based on consensus statements. To illustrate how gaps in basic and clinical knowledge may combine to produce suboptimal disease models on which to base diagnosis and treatment strategies, consider the concept of type I versus type II cardiomyopathy.2 This paradigm was introduced in 2005 to contrast heart failure syndromes resulting from doxorubicin and trastuzumab. …
- Published
- 2017
47. A New Role for the ER Unfolded Protein Response Mediator ATF6
- Author
-
Wendy M. McKimpson and Richard N. Kitsis
- Subjects
0301 basic medicine ,Signal peptide ,Physiology ,Myocardial Reperfusion Injury ,Article ,Rats, Sprague-Dawley ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Myocytes, Cardiac ,Protein disulfide-isomerase ,Peptide sequence ,Mice, Knockout ,biology ,ATF6 ,Myocardium ,Endoplasmic reticulum ,Endoplasmic Reticulum Stress ,Activating Transcription Factor 6 ,Rats ,Mice, Inbred C57BL ,Oxidative Stress ,HEK293 Cells ,030104 developmental biology ,Animals, Newborn ,Biochemistry ,030220 oncology & carcinogenesis ,Chaperone (protein) ,Unfolded protein response ,biology.protein ,Protein folding ,Cardiology and Cardiovascular Medicine ,HeLa Cells ,Signal Transduction - Abstract
Optimal cellular health requires normal protein function, which, in turn, is dependent on proteins folding into their appropriate 3-dimensional conformations. Roughly a third of cellular proteins undergo folding in the lumen of the endoplasmic reticulum (ER). However, under stressful conditions, unfolded proteins sometimes accumulate. This initiates a process termed the unfolded protein response (UPR).1 One arm of the UPR is activated by the ER membrane protein ATF6 (activating transcription factor 6), a fragment of which moves to the nucleus where it functions as a transcription factor to activate genes that mediate protein folding. Unexpectedly, Jin et al2 report in the current issue of Circulation Research a new function for ATF6: to transcriptionally activate the expression of a repertoire of antioxidant genes, many of which encode proteins that reside outside of the ER. Moreover, data are presented supporting a model in which this antioxidant program attenuates myocardial damage from ischemia/reperfusion. Article, see p 862 Some of the information required for protein folding is intrinsic to its amino acid sequence, but chaperones are critical for most proteins to assume their appropriate conformation. Chaperones are a biochemically diverse group of proteins that reside in subcellular compartments where protein folding takes place (eg, cytosol, mitochondria, and ER). Essentially all proteins destined for various cellular membranes and most that will be secreted are directed to the ER by a signal sequence as they exit ribosomes in the cytoplasm. Folding in the ER is coordinated with post-translational modifications that include removal of the signal sequence, N-linked glycosylation, and disulfide bond formation.3 These events require the integrated actions of several classes of chaperones, oxidoreductases (including protein disulfide isomerases), and prolyl peptidyl cis–trans isomerases. When everything proceeds according to plan, high-quality proteins are produced, and the cell is happy. However, things can go awry. Although …
- Published
- 2017
48. Up-regulation of cofilin-1 in cell senescence associates with morphological change and p27
- Author
-
Cheng-Han, Tsai, Chun-Yuan, Chang, Bing-Ze, Lin, Yu-Lou, Wu, Meng-Hsiu, Wu, Liang-Tin, Lin, Wen-Chien, Huang, Jonathan D, Holz, Tzong-Jen, Sheu, Jhih-Shian, Lee, Richard N, Kitsis, Pei-Han, Tai, and Yi-Jang, Lee
- Subjects
Cofilin 1 ,p27Kip1 ,senescence ,cofilin‐1 ,macromolecular substances ,Original Articles ,environment and public health ,Up-Regulation ,growth arrest ,Proliferating Cell Nuclear Antigen ,morphology ,Humans ,Original Article ,TEAD1 ,Cellular Senescence - Abstract
Morphological change is an explicit characteristic of cell senescence, but the underlying mechanisms remains to be addressed. Here, we demonstrated, after a survey of various actin‐binding proteins, that the post‐translational up‐regulation of cofilin‐1 was essential for the reduced rate of actin depolymerization morphological enlargement in senescent cells. Additionally, up‐regulated cofilin‐1 mainly existed in the serine‐3 phosphorylated form, according to the 2D gel immunoblotting assay. The up‐regulation of cofilin‐1 was also detected in aged mammalian tissues. The over‐expression of wild‐type cofilin‐1 and constitutively phosphorylated cofilin‐1 promoted cell senescence with an increased cell size. Additionally, senescent phenotypes were also reduced by knockdown of total cofilin‐1, which led to a decrease in phosphorylated cofilin‐1. The senescence induced by the over‐expression of cofilin‐1 was dependent on p27Kip1, but not on the p53 and p16INK4 expressions. The knockdown of p27Kip1 alleviated cell senescence induced by oxidative stress or replicative stress. We also found that the over‐expression of cofilin‐1 induced the expression of p27Kip1 through transcriptional suppression of the transcriptional enhancer factors domain 1 (TEAD1) transcription factor. The TEAD1 transcription factor played a transrepressive role in the p27Kip1 gene promoter, as determined by the promoter deletion reporter gene assay. Interestingly, the down‐regulation of TEAD1 was accompanied by the up‐regulation of cofilin‐1 in senescence. The knockdown and restoration of TEAD1 in young cells and old cells could induce and inhibit p27Kip1 and senescent phenotypes, respectively. Taken together, the current data suggest that cofilin‐1/TEAD1/p27Kip1 signaling is involved in senescence‐related morphological change and growth arrest., Triggers of senescence, such as replicative stress or oxidative stress, can induce the expression of cofilin‐1 to influence of cell morphology. Up‐regulated cofilin‐1 would enhance the expression of p27kip1 gene by suppressing the TEAD‐1 that transrepresses p27kip1 and lead to senescence‐associated growth delay.
- Published
- 2018
49. Heart disease and cancer – are the two killers colluding?
- Author
-
Sergio Lavandero, Jaime A. Riquelme, and Richard N. Kitsis
- Subjects
Oncology ,medicine.medical_specialty ,biology ,Heart disease ,business.industry ,Cancer ,030204 cardiovascular system & hematology ,medicine.disease ,Fibronectins ,Article ,Transplantation ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Physiology (medical) ,Internal medicine ,Heart failure ,medicine ,biology.protein ,Cardiology and Cardiovascular Medicine ,business ,Ceruloplasmin - Published
- 2018
50. Abstract 117: Regulation of Cardiac Mitochondrial Function by Chaperone Mediated Autophagy
- Author
-
Min Zheng, Jaehoon Lee, Christian Garcia, Nina Kaludercic, Yun Chen, Julio M Matute, Hong Li, Edward Owusu-Ansah, Victor Paulino, Richard N. Kitsis, and Ana Maria Cuervo
- Subjects
Cardiac function curve ,Chaperone-mediated autophagy ,Physiology ,Chemistry ,Autophagy ,Cardiomyopathy ,medicine ,Mitochondrion ,Cardiology and Cardiovascular Medicine ,medicine.disease ,health care economics and organizations ,Function (biology) ,Cell biology - Abstract
Macroautophagy is required for normal cardiac function. Chaperone mediated autophagy (CMA) is distinct in that substrates are transported to lysosomes in complex with Hsc70, rather than within autophagosomes; restricted to cytoplasmic proteins; and selectively marked with a CMA-targeting motif. The role of CMA in the heart has not been assessed. We determined that CMA is induced in the heart by pressure overload with kinetics that differ markedly from those of macroautophagy in response to the same stimulus. In CMA, cargo is imported into the lysosome by LAMP2A, which is necessary, specific, and rate-limiting for this process. Accordingly, we created knockout mice lacking LAMP2A in cardiomyocytes to inactivate CMA specifically in those cells. Unexpectedly, these mice exhibited resistance to stress-induced cardiac dysfunction in both pressure overload and myocardial infarction models. Functional assessment of cardiac mitochondria from L2AKO mice and MEFs exhibited increased rates of O 2 consumption and ATP generation. Mitochondrial proteomics showed increased levels of ATP5L, a component of mitochondrial Complex V (ATP synthase). Overexpression of ATP5L phenocopied LAMP2A KO with respect to augmentation of mitochondrial function. Conversely, knockdown of ATP5L reversed increases in mitochondrial function resulting from L2AKO. We showed that ATP5L, which possesses a KFERQ motif, is a bona fide CMA substrate as it interacts with Hsc70 in a KFERQ-dependent manner; and localizes to lyososmes in a LAMP2A-dependent manner when CMA is induced. ATP5L is thought to mediate oligomerization of Complex V to enhance ATP production. In fact, we observed increases in complex V oligomerization in L2A KO mouse hearts in both basal and TAC-induced states. In summary, cardiomyocyte-specific inhibition of CMA, which renders mice resistant to stress-induced cardiac dysfunction, augments cardiac mitochondrial function through accumulation of ATP5L. Conversely, activation of CMA by cardiac stress may function as a new pathway to heart failure.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.