Compromised mitochondrial quality control triggers lipin1-related rhabdomyolysis

Yamina Hamel, François-Xavier Mauvais, Marine Madrange, Perrine Renard, Corinne Lebreton, Ivan Nemazanyy, Olivier Pellé, Nicolas Goudin, Xiaoyun Tang, Mathieu P Rodero, Caroline Tuchmann-Durand, Patrick Nusbaum, David N Brindley, Peter van Endert, Pascale de Lonlay, Yamina Hamel, François-Xavier Mauvais, Marine Madrange, Perrine Renard, Corinne Lebreton, Ivan Nemazanyy, Olivier Pellé, Nicolas Goudin, Xiaoyun Tang, Mathieu P Rodero, Caroline Tuchmann-Durand, Patrick Nusbaum, David N Brindley, Peter van Endert, Pascale de Lonlay

Abstract

LPIN1 mutations are responsible for inherited recurrent rhabdomyolysis, a life-threatening condition with no efficient therapeutic intervention. Here, we conduct a bedside-to-bench-and-back investigation to study the pathophysiology of lipin1 deficiency. We find that lipin1-deficient myoblasts exhibit a reduction in phosphatidylinositol-3-phosphate close to autophagosomes and late endosomes that prevents the recruitment of the GTPase Armus, locks Rab7 in the active state, inhibits vesicle clearance by fusion with lysosomes, and alters their positioning and function. Oxidized mitochondrial DNA accumulates in late endosomes, where it activates Toll-like receptor 9 (TLR9) and triggers inflammatory signaling and caspase-dependent myolysis. Hydroxychloroquine blocks TLR9 activation by mitochondrial DNA in vitro and may attenuate flares of rhabdomyolysis in 6 patients treated. We suggest a critical role for defective clearance of oxidized mitochondrial DNA that activates TLR9-restricted inflammation in lipin1-related rhabdomyolysis. Interventions blocking TLR9 activation or inflammation can improve patient care in vivo.

Trial registration: ClinicalTrials.gov NCT04007562.

Keywords: Toll-like receptor 9; autophagosome; hydroxychloroquine; inflammation; late endosome; lipin1; mitochondrial quality control; rhabdomyolysis.

Conflict of interest statement

Y.H., F.-X.M., C.L., P.v.E., and P.d.L. have filed PCT (WO/2017/085115; EP3377095; PCT/ EP2016/077843) and US patent applications (US20180325890). Y.H., F.-X.M., M.M., P.v.E., and P.d.L. have filed PCT (WO/2019/020732; PCT/EP2018/070256) patent applications. The remaining authors declare no competing interests.

© 2021 The Authors.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Lipin1 localizes in proximity of late endosomes, autophagosomes, and mitochondria in human myoblasts (A) The confocal images show a representative staining of endogenous lipin1 in primary myoblasts from 1 of 6 controls and 6 patients carrying LPIN1 mutations (Table S1) and in immortalized myoblasts from 1 healthy donor transduced with sh-LPIN1 or control scrambled (sh-Sc) shRNA. (B) Immunoblot analysis of endogenous lipin1 expression as in (A) in 5 patients. (C) Dot plots (mean of 3 technical replicates/dot) depict the means ± SDs of PAP activity quantified in myoblasts from 6 controls and 5 patients (Mann-Whitney U test). (D) Lipin1 distribution within myoblast organelles of 5 controls by confocal microscopy. Dot plots (50–80 cells/dot) show the means ± SDs of the percentage of lipin1 in proximity with a given protein. The images are representative of 1 of at least 3 independent experiments. Scale bars, 10 μm.
Figure 2
Figure 2
Lipin1 deficiency results in a loss of PI3P close to Rab7 structures in myoblasts (A) The confocal images show a representative 2xFYVE-GFP staining from 1 of 6 controls and patients. Dot plots (50 images/dot) show the means ± SDs number of 2xFYVE-GFP dots per patient versus control myoblast (Mann-Whitney U test). (B) As in (A), but using immortalized myoblasts from 1 healthy donor treated with sh-Sc or sh-LPIN1. The plots (30–45 cells/condition) show the number of 2xFYVE-GFP dots (Mann-Whitney U test). (C) Typical distribution of PI3P within the myoblast vesicles from 1 of 3 controls and patients. Box and whisker plots (50 images/condition) depict the percentage of proximity between PI3P and the given marker for 1 representative control and patient (Mann-Whitney U test). (D) Myoblasts from 3 controls and 3 patients were stained for PI35P2 and late endosomal or lysosomal markers. Proximity was calculated as in (C) (unpaired t test). Results are from 1 representative of at least 3 independent experiments. Scale bars, 10 μm. See also Figure S1.
Figure 3
Figure 3
Lipin1-deficient myoblasts display alterations in Rab7 positioning and activation and impaired lysosomal function (A) The distribution of endogenous Rab7 in 30 individual patient and control myoblasts was determined by calculating the intensity of Rab7 fluorescence along a longitudinal axis traversing the nucleus. (B) The interaction between Rab7 and FYCO1 determined by fluorescence resonance energy transfer (FRET) after staining with Rab7 (green) only or for Rab7 (donor) and FYCO1 (acceptor) in red was depicted by line graphs (ns, nanoseconds). (C) Myoblasts transfected with RFP-RILP were immunostained for Rab7. Box and whisker plots (30 images/condition) show the percentage of proximity of Rab7 with RFP-RILP (unpaired t test). (D and E) Myoblasts transfected with a construct encoding RFP-Armus were stained for Rab7 or PI3P. Box and whisker plots (50 images/condition) show the percentage of proximity of Rab7 (D) or PI3P (E) with RFP-Armus (Mann-Whitney U test). (F) Dot plot (mean of 2 technical replicates/dot) with lines depicts the relative level of residual EGFR as compared to β-actin upon incubation with EGF for indicated times. (G) Myoblasts were loaded with DQ-OVA, and fluorescence resulting from the cleavage of DQ-OVA substrate was analyzed by flow cytometry. Dot plots (mean of 2 technical replicates/dot) show the means ± SDs of the value of the mean fluorescence intensity (MFI) of DQ-OVA 60 and 120 min after uptake (mean effect of interaction F(1,6) = 14.43, p = 0.009, of time F(1,6) = 53.46, p = 0.0003, of subjects F(1,6) = 11.23, p = 0.0154). (H) Levels of immature and mature cathepsin D relative to β-actin expression in primary myoblasts were determined by immunoblot. (I) Myoblasts cultured in GM or EBSS were loaded with the Lysotracker Red dye, then stained for endogenous Rab7, LAMP1, and LC3. Box and whisker plots (30 images/condition) show the percentage of proximity of Rab7 (mean effect of interaction F(1,116) = 22.01, p 

Figure 4

Autophagic clearance and mitochondrial quality…

Figure 4

Autophagic clearance and mitochondrial quality and functions are impaired in human lipin1-deficient myoblasts…

Figure 4
Autophagic clearance and mitochondrial quality and functions are impaired in human lipin1-deficient myoblasts (A) Autolysosome formation (red puncta) in myoblasts transfected with the RFP-GFP-LC3 construct upon exposure to EBSS and refed or not with GM. Box and whisker plots (50 images/condition) represent the percentage of autolysosomes (mean effect of interaction F(1,196) = 44.4, p 12S mtDNA levels qPCR in cytosolic fractions from myoblasts of 4 patients and controls exposed to CCCP or EBSS (mean effect of interaction F(1,12) = 0.004867, p = 0.9455, of stimulus F(1,12) = 1.798, p = 0.2047, of subjects F(1,12) = 0.03755, p = 0.8496) and of the mtDNA motif DLOOP (mean effect of interaction F(1,12) = 0.2168, p = 0.6498, of stimulus F(1,12) = 0.3160, p = 0.5844, of subjects F(1,12) = 3.371, p = 0.0913). Dot plots (mean of 3 technical replicates/dot) depict the means ± SDs of the ratio of cycle threshold (CT) values of the given cytosolic fraction normalized to unfractioned cells. Scale bars, 10 μm (A and B). ∗p 

Figure 5

Accumulation of oxidized mitochondria enhances…

Figure 5

Accumulation of oxidized mitochondria enhances inflammation in lipin1-deficient cells through a TLR9 pathway…

Figure 5
Accumulation of oxidized mitochondria enhances inflammation in lipin1-deficient cells through a TLR9 pathway (A) Circulating mtDNA was quantified in plasma by qPCR and inflammatory molecules were measured in sera by flow cytometry, from 12 healthy donors (6 for mtDNA) and 6 patients (Mann-Whitney U test). Dot plots (mean of 3 technical replicates/dot) show the means ± SDs. (B) IFN signature in peripheral blood mononuclear cells was identified by qPCR in 4 controls and 4 patients. Dot plots (mean of 3 technical replicates/dot) show the mean ± SD of the CT value of a given gene normalized to the CT value of BACT (Mann-Whitney U test). (C) DC maturation reflected by CD83 expression (mean effect of interaction F(4,70) = 2.114, p = 0.0881, of stimulus F(4,70) = 36.27, p SIGLEC1 (mean effect of interaction F(1,8) = 4.436, p = 0.0683, of stimulus F(1,8) = 9.255, p = 0.0160, of subjects F(1,8) = 20.78, p = 0.0019), ISG15 (mean effect of interaction F(1,8) = 0.3191, p = 0.5876, of stimulus F(1,8) = 21.75, p = 0.0016, of subjects F(1,8) = 41.33, p = 0.0002), and IFI27 (mean effect of interaction F(1,8) = 21.99, p = 0.0016, of stimulus F(1,8) = 25.45, p = 0.0010, of subjects F(1,8) = 91.20, p < 0.0001) expression. Dot plots (mean of 3 technical replicates/dot) show the means ± SDs of CT value calculated as in (B). (E) Production of IL-6 (ELISA) by myoblasts from 8 controls and 6 patients (mean effect of interaction F(2,36) = 3.882, p = 0.0297, of stimulus F(2,36) = 45.49, p IFNA (mean effect of interaction F(3,24) = 23.44, p < 0.0001, of stimulus F(3,24) = 74.13, p < 0.0001, of subjects F(1,24) = 42.33, p < 0.0001), and IFNB (mean effect of interaction F(3,24) = 2.207, p = 0.1133, of stimulus F(3,24) = 10.19, p = 0.0002, of subjects F(1,24) = 13.60, p = 0.0012). (F) As in (E), but using sh-Sc or sh-LPIN1-transduced immortalized myoblasts from 1 healthy donor (1 different cell vial from the same single healthy donor/dot) and primary myoblasts from 4 patients. Dot plots (mean of 4 technical replicates/dot) show the means ± SDs of the IL-6 concentration (mean effect of interaction F(2,14) = 6.234, p = 0.0116, of stimulus F(1,14) = 0.0874, p = 0.3653, of subjects F(2,14) = 4.397, p = 0.0330). (G) As in (F), but primary myoblasts from 4 controls and patients were transduced with a lentivirus expressing a plasmid encoding for a myc-DDK-tagged lipin1 protein (+LPIN1) or an empty vector (+vector) before being challenged (mean effect of interaction F(2,18) = 0.2196, p = 0.8050, of stimulus F(1,18) = 0.2906, p = 0.5965, of subjects F(2,18) = 14.80, p = 0.0002). (H) Myoblasts from 6 controls and patients were exposed to a vehicle or ethidium bromide (Et. B.). Dot plots (mean of 3 technical replicates/dot) show the means ± SDs of the ratio of the CT value for 12S mitochondrial DNA normalized to the CT value for BACT (mean effect of interaction F(1,12) = 3.186, p = 0.0996, of stimulus F(1,12) = 101.6, p < 0.0001, of subjects F(1,12) = 5.268, p = 0.0405). (I) Myoblasts from 6 controls and patients pre-treated with Et. B. for 5 days were exposed to EBSS and challenged or not with CpG-A for 16 h. Dot plots (mean of 4 technical replicates/dot) show the mean ± SD of IL-6 concentration in culture supernatants at the end of the experiment (mean effect of interaction F(3,24) = 4.989, p = 0.0079, of stimulus F(3,24) = 8.218, p = 0.0006, of subjects F(1,24) = 15.14, p = 0.0007). (J) Myoblasts were transfected with a plasmid encoding HA-tagged TLR9 and exposed to GM or EBSS and with vehicle or CpG-A before refeeding (Refed) cells exposed to EBSS and CpG-A with GM. Cells were then stained for HA-TLR9 (anti-HA antibody, red) and 8OHDG (green). Box whisker plots (25 images/condition) show the percentage of proximity of TLR9 with 8OHDG (mean effect of interaction F(4,240) = 1.712, p = 0.1481, of stimulus F(4,240) = 25.07, p 

Figure 6

mtDNA accumulation is responsible for…

Figure 6

mtDNA accumulation is responsible for inflammation and rhabdomyolysis that can be reversed by…

Figure 6
mtDNA accumulation is responsible for inflammation and rhabdomyolysis that can be reversed by steroids or chloroquine (CLQ) treatment in vivo (A) Real-time calcium flux in myoblasts from 4 controls and patients challenged with CpG-A, after pre-incubation with ODN151, a synthetic TLR9 antagonist. Each dot plot with lines depicts the mean ± SD of the intensity of the fluo-4 calcium probe as normalized to basal intensity of 1 single individual (≥10 cells/condition). (B) Death of myoblasts was evaluated by staining cells from 4 control individuals transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein (+LPIN1) and 4 patients transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein or an empty vector, for the apoptosis marker caspase3/7 green and propidium iodide or 7-actinomycin D (7-AAD), a dye that stains both apoptotic and necrotic cells. Staurosporin (ST): positive control. Fluorescence-activated cell sorting (FACS) plots show the codistribution of the fluorescence intensity of caspase3/7 and of 7-AAD in 1 of 4 controls and patients. (C) Dot plots (≥1,000 single cells/dot) depict the mean ± SD of the proportion of dead cells (ie, cells positive for capsase3/7 and/or 7-AAD) from (B) (mean effect of interaction F(8,45) = 5.296, p = 0.0001, of stimulus F(4,45) = 70.03, p LPIN1 shRNA (mean effect of interaction F(4,20) = 6.154, p = 0.0021, of stimulus F(4,20) = 48.93, p < 0.0001, of subjects F(1,20) = 15.62, p = 0.0008). (E and F) As in (C), but myoblasts were pre-treated with Et. B. or vehicle (mean effect of interaction F(3,24) = 21.28, p 
All figures (7)
Similar articles
Cited by
References
    1. Cervellin G., Comelli I., Lippi G. Rhabdomyolysis: historical background, clinical, diagnostic and therapeutic features. Clin. Chem. Lab. Med. 2010;48:749–756. - PubMed
    1. Luck R.P., Verbin S. Rhabdomyolysis: a review of clinical presentation, etiology, diagnosis, and management. Pediatr. Emerg. Care. 2008;24:262–268. - PubMed
    1. Michot C., Hubert L., Brivet M., De Meirleir L., Valayannopoulos V., Müller-Felber W., Venkateswaran R., Ogier H., Desguerre I., Altuzarra C. LPIN1 gene mutations: a major cause of severe rhabdomyolysis in early childhood. Hum. Mutat. 2010;31:E1564–E1573. - PubMed
    1. Baba T., Kashiwagi Y., Arimitsu N., Kogure T., Edo A., Maruyama T., Nakao K., Nakanishi H., Kinoshita M., Frohman M.A. Phosphatidic acid (PA)-preferring phospholipase A1 regulates mitochondrial dynamics. J. Biol. Chem. 2014;289:11497–11511. - PMC - PubMed
    1. Donkor J., Sariahmetoglu M., Dewald J., Brindley D.N., Reue K. Three mammalian lipins act as phosphatidate phosphatases with distinct tissue expression patterns. J. Biol. Chem. 2007;282:3450–3457. - PubMed
Show all 71 references
Publication types
MeSH terms
Substances
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4
Figure 4
Autophagic clearance and mitochondrial quality and functions are impaired in human lipin1-deficient myoblasts (A) Autolysosome formation (red puncta) in myoblasts transfected with the RFP-GFP-LC3 construct upon exposure to EBSS and refed or not with GM. Box and whisker plots (50 images/condition) represent the percentage of autolysosomes (mean effect of interaction F(1,196) = 44.4, p 12S mtDNA levels qPCR in cytosolic fractions from myoblasts of 4 patients and controls exposed to CCCP or EBSS (mean effect of interaction F(1,12) = 0.004867, p = 0.9455, of stimulus F(1,12) = 1.798, p = 0.2047, of subjects F(1,12) = 0.03755, p = 0.8496) and of the mtDNA motif DLOOP (mean effect of interaction F(1,12) = 0.2168, p = 0.6498, of stimulus F(1,12) = 0.3160, p = 0.5844, of subjects F(1,12) = 3.371, p = 0.0913). Dot plots (mean of 3 technical replicates/dot) depict the means ± SDs of the ratio of cycle threshold (CT) values of the given cytosolic fraction normalized to unfractioned cells. Scale bars, 10 μm (A and B). ∗p 

Figure 5

Accumulation of oxidized mitochondria enhances…

Figure 5

Accumulation of oxidized mitochondria enhances inflammation in lipin1-deficient cells through a TLR9 pathway…

Figure 5
Accumulation of oxidized mitochondria enhances inflammation in lipin1-deficient cells through a TLR9 pathway (A) Circulating mtDNA was quantified in plasma by qPCR and inflammatory molecules were measured in sera by flow cytometry, from 12 healthy donors (6 for mtDNA) and 6 patients (Mann-Whitney U test). Dot plots (mean of 3 technical replicates/dot) show the means ± SDs. (B) IFN signature in peripheral blood mononuclear cells was identified by qPCR in 4 controls and 4 patients. Dot plots (mean of 3 technical replicates/dot) show the mean ± SD of the CT value of a given gene normalized to the CT value of BACT (Mann-Whitney U test). (C) DC maturation reflected by CD83 expression (mean effect of interaction F(4,70) = 2.114, p = 0.0881, of stimulus F(4,70) = 36.27, p SIGLEC1 (mean effect of interaction F(1,8) = 4.436, p = 0.0683, of stimulus F(1,8) = 9.255, p = 0.0160, of subjects F(1,8) = 20.78, p = 0.0019), ISG15 (mean effect of interaction F(1,8) = 0.3191, p = 0.5876, of stimulus F(1,8) = 21.75, p = 0.0016, of subjects F(1,8) = 41.33, p = 0.0002), and IFI27 (mean effect of interaction F(1,8) = 21.99, p = 0.0016, of stimulus F(1,8) = 25.45, p = 0.0010, of subjects F(1,8) = 91.20, p < 0.0001) expression. Dot plots (mean of 3 technical replicates/dot) show the means ± SDs of CT value calculated as in (B). (E) Production of IL-6 (ELISA) by myoblasts from 8 controls and 6 patients (mean effect of interaction F(2,36) = 3.882, p = 0.0297, of stimulus F(2,36) = 45.49, p IFNA (mean effect of interaction F(3,24) = 23.44, p < 0.0001, of stimulus F(3,24) = 74.13, p < 0.0001, of subjects F(1,24) = 42.33, p < 0.0001), and IFNB (mean effect of interaction F(3,24) = 2.207, p = 0.1133, of stimulus F(3,24) = 10.19, p = 0.0002, of subjects F(1,24) = 13.60, p = 0.0012). (F) As in (E), but using sh-Sc or sh-LPIN1-transduced immortalized myoblasts from 1 healthy donor (1 different cell vial from the same single healthy donor/dot) and primary myoblasts from 4 patients. Dot plots (mean of 4 technical replicates/dot) show the means ± SDs of the IL-6 concentration (mean effect of interaction F(2,14) = 6.234, p = 0.0116, of stimulus F(1,14) = 0.0874, p = 0.3653, of subjects F(2,14) = 4.397, p = 0.0330). (G) As in (F), but primary myoblasts from 4 controls and patients were transduced with a lentivirus expressing a plasmid encoding for a myc-DDK-tagged lipin1 protein (+LPIN1) or an empty vector (+vector) before being challenged (mean effect of interaction F(2,18) = 0.2196, p = 0.8050, of stimulus F(1,18) = 0.2906, p = 0.5965, of subjects F(2,18) = 14.80, p = 0.0002). (H) Myoblasts from 6 controls and patients were exposed to a vehicle or ethidium bromide (Et. B.). Dot plots (mean of 3 technical replicates/dot) show the means ± SDs of the ratio of the CT value for 12S mitochondrial DNA normalized to the CT value for BACT (mean effect of interaction F(1,12) = 3.186, p = 0.0996, of stimulus F(1,12) = 101.6, p < 0.0001, of subjects F(1,12) = 5.268, p = 0.0405). (I) Myoblasts from 6 controls and patients pre-treated with Et. B. for 5 days were exposed to EBSS and challenged or not with CpG-A for 16 h. Dot plots (mean of 4 technical replicates/dot) show the mean ± SD of IL-6 concentration in culture supernatants at the end of the experiment (mean effect of interaction F(3,24) = 4.989, p = 0.0079, of stimulus F(3,24) = 8.218, p = 0.0006, of subjects F(1,24) = 15.14, p = 0.0007). (J) Myoblasts were transfected with a plasmid encoding HA-tagged TLR9 and exposed to GM or EBSS and with vehicle or CpG-A before refeeding (Refed) cells exposed to EBSS and CpG-A with GM. Cells were then stained for HA-TLR9 (anti-HA antibody, red) and 8OHDG (green). Box whisker plots (25 images/condition) show the percentage of proximity of TLR9 with 8OHDG (mean effect of interaction F(4,240) = 1.712, p = 0.1481, of stimulus F(4,240) = 25.07, p 

Figure 6

mtDNA accumulation is responsible for…

Figure 6

mtDNA accumulation is responsible for inflammation and rhabdomyolysis that can be reversed by…

Figure 6
mtDNA accumulation is responsible for inflammation and rhabdomyolysis that can be reversed by steroids or chloroquine (CLQ) treatment in vivo (A) Real-time calcium flux in myoblasts from 4 controls and patients challenged with CpG-A, after pre-incubation with ODN151, a synthetic TLR9 antagonist. Each dot plot with lines depicts the mean ± SD of the intensity of the fluo-4 calcium probe as normalized to basal intensity of 1 single individual (≥10 cells/condition). (B) Death of myoblasts was evaluated by staining cells from 4 control individuals transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein (+LPIN1) and 4 patients transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein or an empty vector, for the apoptosis marker caspase3/7 green and propidium iodide or 7-actinomycin D (7-AAD), a dye that stains both apoptotic and necrotic cells. Staurosporin (ST): positive control. Fluorescence-activated cell sorting (FACS) plots show the codistribution of the fluorescence intensity of caspase3/7 and of 7-AAD in 1 of 4 controls and patients. (C) Dot plots (≥1,000 single cells/dot) depict the mean ± SD of the proportion of dead cells (ie, cells positive for capsase3/7 and/or 7-AAD) from (B) (mean effect of interaction F(8,45) = 5.296, p = 0.0001, of stimulus F(4,45) = 70.03, p LPIN1 shRNA (mean effect of interaction F(4,20) = 6.154, p = 0.0021, of stimulus F(4,20) = 48.93, p < 0.0001, of subjects F(1,20) = 15.62, p = 0.0008). (E and F) As in (C), but myoblasts were pre-treated with Et. B. or vehicle (mean effect of interaction F(3,24) = 21.28, p 
All figures (7)
Similar articles
Cited by
References
    1. Cervellin G., Comelli I., Lippi G. Rhabdomyolysis: historical background, clinical, diagnostic and therapeutic features. Clin. Chem. Lab. Med. 2010;48:749–756. - PubMed
    1. Luck R.P., Verbin S. Rhabdomyolysis: a review of clinical presentation, etiology, diagnosis, and management. Pediatr. Emerg. Care. 2008;24:262–268. - PubMed
    1. Michot C., Hubert L., Brivet M., De Meirleir L., Valayannopoulos V., Müller-Felber W., Venkateswaran R., Ogier H., Desguerre I., Altuzarra C. LPIN1 gene mutations: a major cause of severe rhabdomyolysis in early childhood. Hum. Mutat. 2010;31:E1564–E1573. - PubMed
    1. Baba T., Kashiwagi Y., Arimitsu N., Kogure T., Edo A., Maruyama T., Nakao K., Nakanishi H., Kinoshita M., Frohman M.A. Phosphatidic acid (PA)-preferring phospholipase A1 regulates mitochondrial dynamics. J. Biol. Chem. 2014;289:11497–11511. - PMC - PubMed
    1. Donkor J., Sariahmetoglu M., Dewald J., Brindley D.N., Reue K. Three mammalian lipins act as phosphatidate phosphatases with distinct tissue expression patterns. J. Biol. Chem. 2007;282:3450–3457. - PubMed
Show all 71 references
Publication types
MeSH terms
Substances
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 5
Figure 5
Accumulation of oxidized mitochondria enhances inflammation in lipin1-deficient cells through a TLR9 pathway (A) Circulating mtDNA was quantified in plasma by qPCR and inflammatory molecules were measured in sera by flow cytometry, from 12 healthy donors (6 for mtDNA) and 6 patients (Mann-Whitney U test). Dot plots (mean of 3 technical replicates/dot) show the means ± SDs. (B) IFN signature in peripheral blood mononuclear cells was identified by qPCR in 4 controls and 4 patients. Dot plots (mean of 3 technical replicates/dot) show the mean ± SD of the CT value of a given gene normalized to the CT value of BACT (Mann-Whitney U test). (C) DC maturation reflected by CD83 expression (mean effect of interaction F(4,70) = 2.114, p = 0.0881, of stimulus F(4,70) = 36.27, p SIGLEC1 (mean effect of interaction F(1,8) = 4.436, p = 0.0683, of stimulus F(1,8) = 9.255, p = 0.0160, of subjects F(1,8) = 20.78, p = 0.0019), ISG15 (mean effect of interaction F(1,8) = 0.3191, p = 0.5876, of stimulus F(1,8) = 21.75, p = 0.0016, of subjects F(1,8) = 41.33, p = 0.0002), and IFI27 (mean effect of interaction F(1,8) = 21.99, p = 0.0016, of stimulus F(1,8) = 25.45, p = 0.0010, of subjects F(1,8) = 91.20, p < 0.0001) expression. Dot plots (mean of 3 technical replicates/dot) show the means ± SDs of CT value calculated as in (B). (E) Production of IL-6 (ELISA) by myoblasts from 8 controls and 6 patients (mean effect of interaction F(2,36) = 3.882, p = 0.0297, of stimulus F(2,36) = 45.49, p IFNA (mean effect of interaction F(3,24) = 23.44, p < 0.0001, of stimulus F(3,24) = 74.13, p < 0.0001, of subjects F(1,24) = 42.33, p < 0.0001), and IFNB (mean effect of interaction F(3,24) = 2.207, p = 0.1133, of stimulus F(3,24) = 10.19, p = 0.0002, of subjects F(1,24) = 13.60, p = 0.0012). (F) As in (E), but using sh-Sc or sh-LPIN1-transduced immortalized myoblasts from 1 healthy donor (1 different cell vial from the same single healthy donor/dot) and primary myoblasts from 4 patients. Dot plots (mean of 4 technical replicates/dot) show the means ± SDs of the IL-6 concentration (mean effect of interaction F(2,14) = 6.234, p = 0.0116, of stimulus F(1,14) = 0.0874, p = 0.3653, of subjects F(2,14) = 4.397, p = 0.0330). (G) As in (F), but primary myoblasts from 4 controls and patients were transduced with a lentivirus expressing a plasmid encoding for a myc-DDK-tagged lipin1 protein (+LPIN1) or an empty vector (+vector) before being challenged (mean effect of interaction F(2,18) = 0.2196, p = 0.8050, of stimulus F(1,18) = 0.2906, p = 0.5965, of subjects F(2,18) = 14.80, p = 0.0002). (H) Myoblasts from 6 controls and patients were exposed to a vehicle or ethidium bromide (Et. B.). Dot plots (mean of 3 technical replicates/dot) show the means ± SDs of the ratio of the CT value for 12S mitochondrial DNA normalized to the CT value for BACT (mean effect of interaction F(1,12) = 3.186, p = 0.0996, of stimulus F(1,12) = 101.6, p < 0.0001, of subjects F(1,12) = 5.268, p = 0.0405). (I) Myoblasts from 6 controls and patients pre-treated with Et. B. for 5 days were exposed to EBSS and challenged or not with CpG-A for 16 h. Dot plots (mean of 4 technical replicates/dot) show the mean ± SD of IL-6 concentration in culture supernatants at the end of the experiment (mean effect of interaction F(3,24) = 4.989, p = 0.0079, of stimulus F(3,24) = 8.218, p = 0.0006, of subjects F(1,24) = 15.14, p = 0.0007). (J) Myoblasts were transfected with a plasmid encoding HA-tagged TLR9 and exposed to GM or EBSS and with vehicle or CpG-A before refeeding (Refed) cells exposed to EBSS and CpG-A with GM. Cells were then stained for HA-TLR9 (anti-HA antibody, red) and 8OHDG (green). Box whisker plots (25 images/condition) show the percentage of proximity of TLR9 with 8OHDG (mean effect of interaction F(4,240) = 1.712, p = 0.1481, of stimulus F(4,240) = 25.07, p 

Figure 6

mtDNA accumulation is responsible for…

Figure 6

mtDNA accumulation is responsible for inflammation and rhabdomyolysis that can be reversed by…

Figure 6
mtDNA accumulation is responsible for inflammation and rhabdomyolysis that can be reversed by steroids or chloroquine (CLQ) treatment in vivo (A) Real-time calcium flux in myoblasts from 4 controls and patients challenged with CpG-A, after pre-incubation with ODN151, a synthetic TLR9 antagonist. Each dot plot with lines depicts the mean ± SD of the intensity of the fluo-4 calcium probe as normalized to basal intensity of 1 single individual (≥10 cells/condition). (B) Death of myoblasts was evaluated by staining cells from 4 control individuals transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein (+LPIN1) and 4 patients transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein or an empty vector, for the apoptosis marker caspase3/7 green and propidium iodide or 7-actinomycin D (7-AAD), a dye that stains both apoptotic and necrotic cells. Staurosporin (ST): positive control. Fluorescence-activated cell sorting (FACS) plots show the codistribution of the fluorescence intensity of caspase3/7 and of 7-AAD in 1 of 4 controls and patients. (C) Dot plots (≥1,000 single cells/dot) depict the mean ± SD of the proportion of dead cells (ie, cells positive for capsase3/7 and/or 7-AAD) from (B) (mean effect of interaction F(8,45) = 5.296, p = 0.0001, of stimulus F(4,45) = 70.03, p LPIN1 shRNA (mean effect of interaction F(4,20) = 6.154, p = 0.0021, of stimulus F(4,20) = 48.93, p < 0.0001, of subjects F(1,20) = 15.62, p = 0.0008). (E and F) As in (C), but myoblasts were pre-treated with Et. B. or vehicle (mean effect of interaction F(3,24) = 21.28, p 
All figures (7)
Similar articles
Cited by
References
    1. Cervellin G., Comelli I., Lippi G. Rhabdomyolysis: historical background, clinical, diagnostic and therapeutic features. Clin. Chem. Lab. Med. 2010;48:749–756. - PubMed
    1. Luck R.P., Verbin S. Rhabdomyolysis: a review of clinical presentation, etiology, diagnosis, and management. Pediatr. Emerg. Care. 2008;24:262–268. - PubMed
    1. Michot C., Hubert L., Brivet M., De Meirleir L., Valayannopoulos V., Müller-Felber W., Venkateswaran R., Ogier H., Desguerre I., Altuzarra C. LPIN1 gene mutations: a major cause of severe rhabdomyolysis in early childhood. Hum. Mutat. 2010;31:E1564–E1573. - PubMed
    1. Baba T., Kashiwagi Y., Arimitsu N., Kogure T., Edo A., Maruyama T., Nakao K., Nakanishi H., Kinoshita M., Frohman M.A. Phosphatidic acid (PA)-preferring phospholipase A1 regulates mitochondrial dynamics. J. Biol. Chem. 2014;289:11497–11511. - PMC - PubMed
    1. Donkor J., Sariahmetoglu M., Dewald J., Brindley D.N., Reue K. Three mammalian lipins act as phosphatidate phosphatases with distinct tissue expression patterns. J. Biol. Chem. 2007;282:3450–3457. - PubMed
Show all 71 references
Publication types
MeSH terms
Substances
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6
Figure 6
mtDNA accumulation is responsible for inflammation and rhabdomyolysis that can be reversed by steroids or chloroquine (CLQ) treatment in vivo (A) Real-time calcium flux in myoblasts from 4 controls and patients challenged with CpG-A, after pre-incubation with ODN151, a synthetic TLR9 antagonist. Each dot plot with lines depicts the mean ± SD of the intensity of the fluo-4 calcium probe as normalized to basal intensity of 1 single individual (≥10 cells/condition). (B) Death of myoblasts was evaluated by staining cells from 4 control individuals transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein (+LPIN1) and 4 patients transduced with a lentivirus expressing a plasmid encoding for myc-DDK-tagged lipin1 protein or an empty vector, for the apoptosis marker caspase3/7 green and propidium iodide or 7-actinomycin D (7-AAD), a dye that stains both apoptotic and necrotic cells. Staurosporin (ST): positive control. Fluorescence-activated cell sorting (FACS) plots show the codistribution of the fluorescence intensity of caspase3/7 and of 7-AAD in 1 of 4 controls and patients. (C) Dot plots (≥1,000 single cells/dot) depict the mean ± SD of the proportion of dead cells (ie, cells positive for capsase3/7 and/or 7-AAD) from (B) (mean effect of interaction F(8,45) = 5.296, p = 0.0001, of stimulus F(4,45) = 70.03, p LPIN1 shRNA (mean effect of interaction F(4,20) = 6.154, p = 0.0021, of stimulus F(4,20) = 48.93, p < 0.0001, of subjects F(1,20) = 15.62, p = 0.0008). (E and F) As in (C), but myoblasts were pre-treated with Et. B. or vehicle (mean effect of interaction F(3,24) = 21.28, p 
All figures (7)

References

    1. Cervellin G., Comelli I., Lippi G. Rhabdomyolysis: historical background, clinical, diagnostic and therapeutic features. Clin. Chem. Lab. Med. 2010;48:749–756.
    1. Luck R.P., Verbin S. Rhabdomyolysis: a review of clinical presentation, etiology, diagnosis, and management. Pediatr. Emerg. Care. 2008;24:262–268.
    1. Michot C., Hubert L., Brivet M., De Meirleir L., Valayannopoulos V., Müller-Felber W., Venkateswaran R., Ogier H., Desguerre I., Altuzarra C. LPIN1 gene mutations: a major cause of severe rhabdomyolysis in early childhood. Hum. Mutat. 2010;31:E1564–E1573.
    1. Baba T., Kashiwagi Y., Arimitsu N., Kogure T., Edo A., Maruyama T., Nakao K., Nakanishi H., Kinoshita M., Frohman M.A. Phosphatidic acid (PA)-preferring phospholipase A1 regulates mitochondrial dynamics. J. Biol. Chem. 2014;289:11497–11511.
    1. Donkor J., Sariahmetoglu M., Dewald J., Brindley D.N., Reue K. Three mammalian lipins act as phosphatidate phosphatases with distinct tissue expression patterns. J. Biol. Chem. 2007;282:3450–3457.
    1. Pascual F., Carman G.M. Phosphatidate phosphatase, a key regulator of lipid homeostasis. Biochim. Biophys. Acta. 2013;1831:514–522.
    1. Sasser T., Qiu Q.S., Karunakaran S., Padolina M., Reyes A., Flood B., Smith S., Gonzales C., Fratti R.A. Yeast lipin 1 orthologue pah1p regulates vacuole homeostasis and membrane fusion. J. Biol. Chem. 2012;287:2221–2236.
    1. Rashid T., Nemazanyy I., Paolini C., Tatsuta T., Crespin P., de Villeneuve D., Brodesser S., Benit P., Rustin P., Baraibar M.A. Lipin1 deficiency causes sarcoplasmic reticulum stress and chaperone-responsive myopathy. EMBO J. 2019;38:e99576.
    1. Schweitzer G.G., Collier S.L., Chen Z., McCommis K.S., Pittman S.K., Yoshino J., Matkovich S.J., Hsu F.F., Chrast R., Eaton J.M. Loss of lipin 1-mediated phosphatidic acid phosphohydrolase activity in muscle leads to skeletal myopathy in mice. FASEB J. 2019;33:652–667.
    1. Michot C., Mamoune A., Vamecq J., Viou M.T., Hsieh L.S., Testet E., Lainé J., Hubert L., Dessein A.F., Fontaine M. Combination of lipid metabolism alterations and their sensitivity to inflammatory cytokines in human lipin-1-deficient myoblasts. Biochim. Biophys. Acta. 2013;1832:2103–2114.
    1. Pelosi M., Testet E., Le Lay S., Dugail I., Tang X., Mabilleau G., Hamel Y., Madrange M., Blanc T., Odent T. Normal human adipose tissue functions and differentiation in patients with biallelic LPIN1 inactivating mutations. J. Lipid Res. 2017;58:2348–2364.
    1. Finck B.N., Gropler M.C., Chen Z., Leone T.C., Croce M.A., Harris T.E., Lawrence J.C., Jr., Kelly D.P. Lipin 1 is an inducible amplifier of the hepatic PGC-1alpha/PPARalpha regulatory pathway. Cell Metab. 2006;4:199–210.
    1. Peterson T.R., Sengupta S.S., Harris T.E., Carmack A.E., Kang S.A., Balderas E., Guertin D.A., Madden K.L., Carpenter A.E., Finck B.N., Sabatini D.M. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell. 2011;146:408–420.
    1. Zhang P., Verity M.A., Reue K. Lipin-1 regulates autophagy clearance and intersects with statin drug effects in skeletal muscle. Cell Metab. 2014;20:267–279.
    1. Meana C., Peña L., Lordén G., Esquinas E., Guijas C., Valdearcos M., Balsinde J., Balboa M.A. Lipin-1 integrates lipid synthesis with proinflammatory responses during TLR activation in macrophages. J. Immunol. 2014;193:4614–4622.
    1. Bergounioux J., Brassier A., Rambaud C., Bustarret O., Michot C., Hubert L., Arnoux J.B., Laquerriere A., Bekri S., Galene-Gromez S. Fatal rhabdomyolysis in 2 children with LPIN1 mutations. J. Pediatr. 2012;160:1052–1054.
    1. Kim H.B., Kumar A., Wang L., Liu G.H., Keller S.R., Lawrence J.C., Jr., Finck B.N., Harris T.E. Lipin 1 represses NFATc4 transcriptional activity in adipocytes to inhibit secretion of inflammatory factors. Mol. Cell. Biol. 2010;30:3126–3139.
    1. Hyttinen J.M., Niittykoski M., Salminen A., Kaarniranta K. Maturation of autophagosomes and endosomes: a key role for Rab7. Biochim. Biophys. Acta. 2013;1833:503–510.
    1. Bissa B., Deretic V. Autophagosome Formation: Cutting the Gordian Knot at the ER. Curr. Biol. 2018;28:R347–R349.
    1. Schink K.O., Tan K.W., Stenmark H. Phosphoinositides in Control of Membrane Dynamics. Annu. Rev. Cell Dev. Biol. 2016;32:143–171.
    1. Jaber N., Mohd-Naim N., Wang Z., DeLeon J.L., Kim S., Zhong H., Sheshadri N., Dou Z., Edinger A.L., Du G. Vps34 regulates Rab7 and late endocytic trafficking through recruitment of the GTPase-activating protein Armus. J. Cell Sci. 2016;129:4424–4435.
    1. Hong Z., Pedersen N.M., Wang L., Torgersen M.L., Stenmark H., Raiborg C. PtdIns3P controls mTORC1 signaling through lysosomal positioning. J. Cell Biol. 2017;216:4217–4233.
    1. Krauß M., Haucke V. A grab to move on: ER-endosome contacts in membrane protrusion formation and neurite outgrowth. EMBO J. 2015;34:1442–1444.
    1. Pankiv S., Alemu E.A., Brech A., Bruun J.A., Lamark T., Overvatn A., Bjørkøy G., Johansen T. FYCO1 is a Rab7 effector that binds to LC3 and PI3P to mediate microtubule plus end-directed vesicle transport. J. Cell Biol. 2010;188:253–269.
    1. Lemmon M.A. Phosphoinositide recognition domains. Traffic. 2003;4:201–213.
    1. Kümmel D., Ungermann C. Principles of membrane tethering and fusion in endosome and lysosome biogenesis. Curr. Opin. Cell Biol. 2014;29:61–66.
    1. Guerra F., Bucci C. Multiple Roles of the Small GTPase Rab7. Cells. 2016;5:E34.
    1. De Luca M., Bucci C. A new V-ATPase regulatory mechanism mediated by the Rab interacting lysosomal protein (RILP) Commun. Integr. Biol. 2014;7:e971572.
    1. Cabukusta B., Neefjes J. Mechanisms of lysosomal positioning and movement. Traffic. 2018;19:761–769.
    1. Pankiv S., Johansen T. FYCO1: linking autophagosomes to microtubule plus end-directing molecular motors. Autophagy. 2010;6:550–552.
    1. Carroll B., Mohd-Naim N., Maximiano F., Frasa M.A., McCormack J., Finelli M., Thoresen S.B., Perdios L., Daigaku R., Francis R.E. The TBC/RabGAP Armus coordinates Rac1 and Rab7 functions during autophagy. Dev. Cell. 2013;25:15–28.
    1. Wang K., Klionsky D.J. Mitochondria removal by autophagy. Autophagy. 2011;7:297–300.
    1. Ugrankar R., Liu Y., Provaznik J., Schmitt S., Lehmann M. Lipin is a central regulator of adipose tissue development and function in Drosophila melanogaster. Mol. Cell. Biol. 2011;31:1646–1656.
    1. West A.P., Shadel G.S. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat. Rev. Immunol. 2017;17:363–375.
    1. Caielli S., Athale S., Domic B., Murat E., Chandra M., Banchereau R., Baisch J., Phelps K., Clayton S., Gong M. Oxidized mitochondrial nucleoids released by neutrophils drive type I interferon production in human lupus. J. Exp. Med. 2016;213:697–713.
    1. Roers A., Hiller B., Hornung V. Recognition of Endogenous Nucleic Acids by the Innate Immune System. Immunity. 2016;44:739–754.
    1. Ohto U., Ishida H., Shibata T., Sato R., Miyake K., Shimizu T. Toll-like Receptor 9 Contains Two DNA Binding Sites that Function Cooperatively to Promote Receptor Dimerization and Activation. Immunity. 2018;48:649–658.e4.
    1. Fang C., Wei X., Wei Y. Mitochondrial DNA in the regulation of innate immune responses. Protein Cell. 2016;7:11–16.
    1. Torres P.A., Helmstetter J.A., Kaye A.M., Kaye A.D. Rhabdomyolysis: pathogenesis, diagnosis, and treatment. Ochsner J. 2015;15:58–69.
    1. Jordens I., Fernandez-Borja M., Marsman M., Dusseljee S., Janssen L., Calafat J., Janssen H., Wubbolts R., Neefjes J. The Rab7 effector protein RILP controls lysosomal transport by inducing the recruitment of dynein-dynactin motors. Curr. Biol. 2001;11:1680–1685.
    1. Saridaki T., Nippold M., Dinter E., Roos A., Diederichs L., Fensky L., Schulz J.B., Falkenburger B.H. FYCO1 mediates clearance of α-synuclein aggregates through a Rab7-dependent mechanism. J. Neurochem. 2018;146:474–492.
    1. Johnson D.E., Ostrowski P., Jaumouillé V., Grinstein S. The position of lysosomes within the cell determines their luminal pH. J. Cell Biol. 2016;212:677–692.
    1. Nakamura S., Yoshimori T. New insights into autophagosome-lysosome fusion. J. Cell Sci. 2017;130:1209–1216.
    1. Filimonenko M., Isakson P., Finley K.D., Anderson M., Jeong H., Melia T.J., Bartlett B.J., Myers K.M., Birkeland H.C., Lamark T. The selective macroautophagic degradation of aggregated proteins requires the PI3P-binding protein Alfy. Mol. Cell. 2010;38:265–279.
    1. Napoli E., Song G., Panoutsopoulos A., Riyadh M.A., Kaushik G., Halmai J., Levenson R., Zarbalis K.S., Giulivi C. Beyond autophagy: a novel role for autism-linked Wdfy3 in brain mitophagy. Sci. Rep. 2018;8:11348.
    1. Cullup T., Kho A.L., Dionisi-Vici C., Brandmeier B., Smith F., Urry Z., Simpson M.A., Yau S., Bertini E., McClelland V. Recessive mutations in EPG5 cause Vici syndrome, a multisystem disorder with defective autophagy. Nat. Genet. 2013;45:83–87.
    1. Hori I., Otomo T., Nakashima M., Miya F., Negishi Y., Shiraishi H., Nonoda Y., Magara S., Tohyama J., Okamoto N. Defects in autophagosome-lysosome fusion underlie Vici syndrome, a neurodevelopmental disorder with multisystem involvement. Sci. Rep. 2017;7:3552.
    1. MacVicar T., Ohba Y., Nolte H., Mayer F.C., Tatsuta T., Sprenger H.G., Lindner B., Zhao Y., Li J., Bruns C. Lipid signalling drives proteolytic rewiring of mitochondria by YME1L. Nature. 2019;575:361–365.
    1. Kameoka S., Adachi Y., Okamoto K., Iijima M., Sesaki H. Phosphatidic Acid and Cardiolipin Coordinate Mitochondrial Dynamics. Trends Cell Biol. 2018;28:67–76.
    1. Sugiura A., McLelland G.L., Fon E.A., McBride H.M. A new pathway for mitochondrial quality control: mitochondrial-derived vesicles. EMBO J. 2014;33:2142–2156.
    1. Park B., Brinkmann M.M., Spooner E., Lee C.C., Kim Y.M., Ploegh H.L. Proteolytic cleavage in an endolysosomal compartment is required for activation of Toll-like receptor 9. Nat. Immunol. 2008;9:1407–1414.
    1. Oka T., Hikoso S., Yamaguchi O., Taneike M., Takeda T., Tamai T., Oyabu J., Murakawa T., Nakayama H., Nishida K. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature. 2012;485:251–255.
    1. Zhang Q., Raoof M., Chen Y., Sumi Y., Sursal T., Junger W., Brohi K., Itagaki K., Hauser C.J. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464:104–107.
    1. Shintani Y., Kapoor A., Kaneko M., Smolenski R.T., D’Acquisto F., Coppen S.R., Harada-Shoji N., Lee H.J., Thiemermann C., Takashima S. TLR9 mediates cellular protection by modulating energy metabolism in cardiomyocytes and neurons. Proc. Natl. Acad. Sci. USA. 2013;110:5109–5114.
    1. Yao M., Liu X., Li D., Chen T., Cai Z., Cao X. Late endosome/lysosome-localized Rab7b suppresses TLR9-initiated proinflammatory cytokine and type I IFN production in macrophages. J. Immunol. 2009;183:1751–1758.
    1. Akbar M.A., Mandraju R., Tracy C., Hu W., Pasare C., Krämer H. ARC Syndrome-Linked Vps33B Protein Is Required for Inflammatory Endosomal Maturation and Signal Termination. Immunity. 2016;45:267–279.
    1. Kok B.P., Kienesberger P.C., Dyck J.R., Brindley D.N. Relationship of glucose and oleate metabolism to cardiac function in lipin-1 deficient (fld) mice. J. Lipid Res. 2012;53:105–118.
    1. Senft D., Ronai Z.A. UPR, autophagy, and mitochondria crosstalk underlies the ER stress response. Trends Biochem. Sci. 2015;40:141–148.
    1. Liu Y., Nguyen P.T., Wang X., Zhao Y., Meacham C.E., Zou Z., Bordieanu B., Johanns M., Vertommen D., Wijshake T. TLR9 and beclin 1 crosstalk regulates muscle AMPK activation in exercise. Nature. 2020;578:605–609.
    1. Mauthe M., Orhon I., Rocchi C., Zhou X., Luhr M., Hijlkema K.J., Coppes R.P., Engedal N., Mari M., Reggiori F. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy. 2018;14:1435–1455.
    1. Chaanine A.H., Gordon R.E., Nonnenmacher M., Kohlbrenner E., Benard L., Hajjar R.J. High-dose chloroquine is metabolically cardiotoxic by inducing lysosomes and mitochondria dysfunction in a rat model of pressure overload hypertrophy. Physiol. Rep. 2015;3:e12413.
    1. Kuznik A., Bencina M., Svajger U., Jeras M., Rozman B., Jerala R. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J. Immunol. 2011;186:4794–4804.
    1. Sacre K., Criswell L.A., McCune J.M. Hydroxychloroquine is associated with impaired interferon-alpha and tumor necrosis factor-alpha production by plasmacytoid dendritic cells in systemic lupus erythematosus. Arthritis Res. Ther. 2012;14:R155.
    1. Torigoe M., Sakata K., Ishii A., Iwata S., Nakayamada S., Tanaka Y. Hydroxychloroquine efficiently suppresses inflammatory responses of human class-switched memory B cells via Toll-like receptor 9 inhibition. Clin. Immunol. 2018;195:1–7.
    1. Hoene V., Peiser M., Wanner R. Human monocyte-derived dendritic cells express TLR9 and react directly to the CpG-A oligonucleotide D19. J. Leukoc. Biol. 2006;80:1328–1336.
    1. Ketel K., Krauss M., Nicot A.S., Puchkov D., Wieffer M., Müller R., Subramanian D., Schultz C., Laporte J., Haucke V. A phosphoinositide conversion mechanism for exit from endosomes. Nature. 2016;529:408–412.
    1. Kimura S., Noda T., Yoshimori T. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy. 2007;3:452–460.
    1. Jongsma M.L., Bakker J., Cabukusta B., Liv N., van Elsland D., Fermie J. SKIP-HOPS recruits TBC1D15 for a Rab7-to-Arl8b identity switch to control late endosome transport. EMBO J. 2020;39 doi: 10.15252/embj.2019102301.
    1. Hamel Y., Blake N., Gabrielsson S., Haigh T., Jooss K., Martinache C., Caillat-Zucman S., Rickinson A.B., Hacein-Bey S., Fischer A., Cavazzana-Calvo M. Adenovirally transduced dendritic cells induce bispecific cytotoxic T lymphocyte responses against adenovirus and cytomegalovirus pp65 or against adenovirus and Epstein-Barr virus EBNA3C protein: a novel approach for immunotherapy. Hum. Gene Ther. 2002;13:855–866.
    1. Schindelin J., Arganda-Carreras I., Frise E., Kaynig V., Longair M., Pietzsch T., Preibisch S., Rueden C., Saalfeld S., Schmid B. Fiji: an open-source platform for biological-image analysis. Nat. Methods. 2012;9:676–682.
    1. Kenworthy A.K., Edidin M. Imaging fluorescence resonance energy transfer as probe of membrane organization and molecular associations of GPI-anchored proteins. Methods Mol. Biol. 1999;116:37–49.

Source: PubMed

3
Abonneren