Endoplasmic reticulum-mitochondrial crosstalk: a novel role for the mitochondrial peptide humanin

2017-03-30 04:44ParameswaranSreekumarDavidHintonRamKannan

Parameswaran G. Sreekumar, David R. Hinton, Ram Kannan,

1 Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA, USA

2 Department of Pathology and Ophthalmology, USC Roski Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA

Endoplasmic reticulum-mitochondrial crosstalk: a novel role for the mitochondrial peptide humanin

Parameswaran G. Sreekumar1, David R. Hinton2,*,#, Ram Kannan1,#

1 Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA, USA

2 Department of Pathology and Ophthalmology, USC Roski Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA

How to cite this article:Sreekumar PG, Hinton DR, Kannan R (2017) Endoplasmic reticulum-mitochondrial crosstalk: a novel role for the mitochondrial peptide humanin. Neural Regen Res 12(1):35-38.

Open access statement:is is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

In this review, the interactive mechanisms of mitochondria with the endoplasmic reticulum (ER) are dis‐cussed with emphasis on the potential protective role of the mitochondria derived peptide humanin (HN) in ER stress.e ER and mitochondria are dynamic organelles capable of modifying their structure and function in response to changing environmental conditions. The ER and mitochondria join together at multiple sites and form mitochondria‐ER associated membranes that participate in signal transduction pathways that are under active investigation. Our laboratory previously showed that HN protects cells from oxidative stress induced cell death and more recently, described the bene fi cial role of HN on ER stress‐in‐duced apoptosis in retinal pigment epithelium cells and the involvement of ER‐mitochondrial cross‐talk in cellular protection.e protection was achieved, in part, by the restoration of mitochondrial glutathione that was depleted by ER stress.us, HN may be a promising candidate for therapy for diseases that involve both oxidative and ER stress. Developing novel approaches for retinal delivery of HN, its analogues as well as small molecular weight ER stress inhibitors would prove to be a valuable approach in the treatment of age‐related macular degeneration.

endoplasmic reticulum; mitochondria; mitochondrial-derived peptide; antioxidants; retinal pigment epithelium; age-related macular degeneration

Accepted: 2017-01-16

Introduction

As one of the largest organelles in eukaryotic cells, the en‐doplasmic reticulum (ER) is a membrane bound network of branching tubules and fl attened sacs that plays a major role in the synthesis, folding, and structural maturation of pro‐teins, especially those destined for secretion or to the plas‐ma membrane (Oakes and Papa, 2015). Multiple cellular stresses, such as oxidative stress, altered protein glycosyla‐tion, or protein folding defects, lead to accumulation of un‐folded or misfolded proteins in the ER lumen and disturb ER function causing “ER‐stress”. These ER‐stress signals activate transcriptional and translational pathways that deal with unfolded and misfolded proteins, known as the un‐folded protein response (UPR). Abnormally folded proteins in the ER can be flushed out through the ER‐associated protein degradation (ERAD) pathway, in which misfolded proteins are translocated to the cytosol, where they undergo ubiquitylation and proteasome‐mediated degradation. Fur‐ther, UPR signaling promotes autophagy, which operates as an active mechanism to eliminate protein aggregates and damaged organellesviathe lysosomal pathway (Oakes and Papa, 2015).

Recent studies have revealed the signi fi cance of ER‐mito‐ chondrial crosstalk in pathophysiological situations.e ER and mitochondria join together at several contact sites to form speci fi c domains, termed mitochondria‐ER associated membranes (MAMs) or mitochondria‐associated ER mem‐branes (MERCs) (Giacomello and Pellegrini, 2016).ese contact sites help them to reciprocally transmit signals and communications with one another under stress conditions, triggering multiple, synergistic responses (Marchi et al., 2014). The roles played by the ER‐mitochondria interface range from the coordination of calcium transfer to the reg‐ulation of mitochondrial fi ssion, autophagy, accumulation of reactive oxygen species (ROS) and inflammasome for‐mation.e relationship between the ER‐mitochondria in‐terface and in fl ammation was revealed with the observation that ROS promote the activation of NLRP3 in fl ammasomes which serve as a platform for caspase 1 activation (Bronner et al., 2015). Overall, it is revealed that the ER‐mitochon‐dria connection plays a fundamental role in the regulation of mitochondrial dynamics (Marchi et al., 2014).

ER Stress in Retinal Disease

A growing number of reports have suggested that accumu‐lation of misfolded proteins plays an important role in thepathogenesis of several degenerative eye diseases such as retinitis pigmentosa, glaucoma, and age‐related macular degeneration (AMD) (Salminen et al., 2010; Zhang et al., 2014). Even though there is no de fi nitive evidence that ER stress is directly involved in AMD pathogenesis, oxidative stress, in fl ammation, cell death and angiogenesis are closely linked with ER stress and AMD (Salminen et al., 2010). For example, high concentrations of oxidized lipids that accu‐mulate in the extracellular deposits found in AMD (drusen) could stimulate vascular endothelial growth factor (VEGF)viaPERK/ATF4 signaling pathway and activate in fl ammato‐ry stimuli which could trigger ER stress (Zhang et al., 2014). It has been proposed that misfolded protein‐induced ER stress in retinal pigment epithelium (RPE) and/or choroid could lead to chronic oxidative stress, complement deregula‐tion and AMD (reviewed in Zhang et al., 2014). Work from our laboratory demonstrated the involvement of ER stress in the regulation of cell death in RPE cells, the site of the early primary pathology in AMD (Dou et al., 2012). Induction of ER stress in the retina and RPE/choroid complex from mice exposed to cigarette smoke, a risk factor for AMD has been reported (Zhang et al., 2014). ER stress induced angiogen‐esis by up‐regulating VEGFviaUPR pathway and down regulating the antiangiogenic pigment epithelium‐derived factor (PEDF) and promoting choroidal neovascularization (Salminen et al., 2010). Further, some ER inhibitors such as sterculic acid are reported to reduce in fl ammation and CNV formation (Zhang et al., 2014).

Humanin is a mitochondria‐derived 21–24 amino acid pep‐tide encoded within the mitochondrial DNA that was first discovered through a search for neuroprotective factors from a cDNA library constructed from an una ff ected brain frac‐tion of an Alzheimer patient (Yen et al., 2013). Subsequently, multiple studies demonstrated its neuroprotective, anti‐in‐flammatory, antiapoptotic, antiaging and antifibrilogenic properties in various cells and tissues (Yen et al., 2013). We have recently provided evidence that HN protected human RPE cells from oxidative cell death (Sreekumar et al., 2016). We found that HN exerted its protective function by utilizing intracellular and extracellular pathways involving mitochon‐dria as well as STAT‐3 signaling (Sreekumar et al., 2016).

Humanin Protection from ER Stress

The study by Matsunaga et al. (2016) raises interesting possibilities to further our understanding of ER stress re‐lated mechanisms in the RPE and retina and potential roles of mitochondrial peptides in cellular protection. As alluded to earlier, chronic ER stress has been linked to an array of neurodegenerative diseases and inhibition of ER stress may provide a novel and effective therapeutic approach in the treatment of retinal diseases. In particular, small molecule inhibitors of the ER pathways could be potential drugs. However, targeting only one branch of the ER stress pathway might not be sufficient to prevent cell death giv‐en the close interactionviamitochondria‐ER associated membranes (Giacomello and Pellegrini, 2016). Therefore, it is ideal to select drugs or agents that target both of these organelles for an effective therapy. Given the multipotent e ff ects of HN in protecting against mitochondrial and ER stress, HN and its analogs could prove to be a valuable therapeutic approach for AMD especially since HN analogs have been reported to be several fold more potent than HN (Yen et al., 2013). HN being a short chain, low molecular peptide, has rapid tissue clearance resulting in less avail‐ability and hence may require frequent administrations. In order to overcome this problem, and to improve retention time, one approach would be to utilize thermally responsive elastin‐like polypeptides (ELP) (Wang et al., 2014) recom‐binantly fused with HN.

Humanin and ER-Mitochondrial Crosstalk

Figure 1 Uptake and translocation of FIC-labeled HN peptide in human RPE cells.

Figure 2 Schematic representation of the ER stress induced by tunicamycin leading to apoptosis in RPE and the protective e ff ect of exogenous Humanin on RPE apoptosis.

With respect to mechanism of HN action in mitochondria, it would be of interest to study factors that a ff ect ER‐induced mitochondrial dysfunction and the pathways involved in this process. However, the localization of endogenous HN in subcellular compartments and sites of synthesis remain a topic of discussion. HN is expressed from an open reading frame (ORF) within the mitochondrial 16S ribosomal RNA. It is encoded from a 75 bp ORF sequence within the 1,567 bp cDNA, which yields either a 21 or 24 amino acid depend‐ing on the location of translation machinery. HN could also be encoded within one or more of the nuclear regions with 92% and 95% similarity to the original HN cDNA dispersed in multiple copies throughout the human genome. It is not clear whether HN is translated in the mitochondria (21‐ami‐no acid peptide) or the cytoplasm (24 amino acid peptide). Using fl uorescein labeled HN and a mitotracker, we obtained evidence that there is rapid and abundant uptake of HN by RPE mitochondria using confocal microscopy (Figure 1).ese experiments also showed the presence of HN in the cytoplasm under the experimental conditions (Figure 1). However, it is not known whether HN enters the ER compartment and whether it is found on the ER surface or is internalized. Thus, additional studies in unstressed and ER‐stressed RPE will be needed to settle that question. Secondly, how ER stressors affect mitochondrial respira‐tion and biogenesis in RPE and the nature of the salutary e ff ects of HN will be an important issue to address. In this context, we showed recently that oxidative stress‐induced decrease in mitochondrial bioenergetics and decrease in mitochondrial DNA copy number in human RPE cells was prevented by HN co‐treatment (Sreekumar et al., 2016). Further, inhibition of the translocation of Bax from cytosolto mitochondria by HN was also observed (Sreekumar et al., 2016).e modulatory e ff ect of ER stress on pro‐ and anti‐apoptotic factors in RPE such as Bax, Bcl‐2 and the e ff ect of HN on these factors remains to be examined. Fi‐nally, the role of mitochondrial HN on mitochondrial‐ER crosstalk mediated through MAMs (e.g. calcium signaling) should be evaluated.

Based on the information available so far, a scheme for ER stress‐induced mitochondrial dysfunction and protec‐tion by HN can be presented (Figure 2). According to this scheme, treatment of RPE by an ER stressor such as TM causes activation of caspase 3 resulting from the activa‐tion of ER‐resident caspase 4 that eventually results in cell death.ere is also a depletion of the antioxidant GSH in mitochondria with ER stress. In ER stress, exogenous HN rapidly enters mitochondria and protects RPE from apop‐totic cell death by upregulating the rate‐limiting enzyme of GSH biosynthesis and increasing mitochondrial GSH levels (Figure 2).

Does the ER‐induced oxidative stress involve mitochon‐drial inflammatory response such as the activation of in‐flammasomeviaNLRP3 and how does HN mediate this process? This question is important since stimulation of in fl ammatory cytokines with oxidative stress is well known and involvement of the in fl ammasomal component NLRP3 in AMD has been described. Further, a mechanism inte‐grating cellular stress with innate immunity was reported in an infection model of ER stress sensor IRE1α (Bronner et al., 2015).

Humanin and Glutathione

As stated earlier, exposure of RPE to TM attenuated mi‐tochondrial GSH and HN restored mitochondrial GSH levels suggesting that HN may be acting at least in part by upregulation of antioxidant enzymes. Glutathione (GSH) is a critical component of mitochondrial antioxidant defense system. GSH synthesis occurs exclusively in cytosol and mi‐tochondria do not possess the synthetic machinery for GSH. However, GSH is found mostly in intracellular organelles in‐cluding mitochondria and ER to maintain organelle‐speci fi c functions and cell survival. Specific carriers for import of GSH from cytosol to mitochondria are known (Ribas et al., 2014), whether HN activates the mitochondria speci fi c GSH carriers in RPE cells remains to be determined.

Conclusions

A better understanding of the mechanisms that orchestrate the ER stress responses and its crosstalk with mitochondria may help to devise future strategies of safely modulating this process for therapeutic bene fi t in retinal neurodegenerative diseases such as AMD, and HN seems to be a promising candidate in this regard.

Acknowledgments:We thank Ernesto Barron for assistance in preparation of the fi gures.

Author contributions:All authors (PGS, DRH, RK) wrote the manuscript and prepared the fi gures.

Con fl icts of interest:None declared.

Bronner DN, Abuaita BH, Chen X, Fitzgerald KA, Nuñez G, He Y, Yin XM, O’Riordan MX (2015) Endoplasmic reticulum stress activates the in fl ammasome via NLRP3‐ and caspase‐2‐driven mitochondrial dam‐age. Immunity 43:451‐462.

Dou G, Sreekumar PG, Spee C, He S, Ryan SJ, Kannan R, Hinton DR (2012) De fi ciency of αB crystallin augments ER stress‐induced apop‐tosis by enhancing mitochondrial dysfunction. Free Radic Biol Med 53:1111‐1122.

Giacomello M, Pellegrini (2016) The coming of age of the mitochon‐dria‐ER contact: a matter of thickness. Cell Death Di ff 23:1417‐1427.

Marchi S, Patergnani S, Pinton P (2014)e endoplasmic reticulum‐mi‐tochondria connection: One touch, multiple functions. Biochim Bio‐phys Acta 1837:461‐469.

Matsunaga D, Sreekumar PG, Ishikawa K, Terasaki H, Barron E, Cohen P, Kannan R, Hinton DR (2016) Humanin protects RPE cells from endoplasmic reticulum stress‐induced apoptosis by upregulation of mitochondrial glutathione. PLoS One 11:e0165150.

Ribas V, Garcia‐Ruiz C, Fernandez‐Checa JC (2014) Glutathione and mitochondria. Front Pharmacol 5:151.

Salminen A, Kauppinen A, Hyttinen JM, Toropainen E, Kaarniranta K (2010) Endoplasmic reticulum stress in age‐related macular degenera‐tion: trigger for neovascularization. Mol Med 16:535‐542.

Sreekumar PG, Ishikawa K, Spee C, Mehta HH, Wan J, Yen K, Cohen P, Kannan R, Hinton DR (2016)e mitochondrial‐derived peptide humanin protects RPE cells from oxidative stress, senescence, and mi‐tochondrial dysfunction. Invest Ophthalmol Vis Sci 57:1238‐1253.

Wang W, Sreekumar PG, Valluripalli V, Shi P, Wang J, Lin YA, Cui H, Kannan R, Hinton DR, MacKay JA (2014) Protein polymer nanopar‐ticles engineered as chaperones protect against apoptosis in human retinal pigment epithelial cells. J Control Release 191:4‐14.

Yen K, Lee C, Mehta H, Cohen P (2013)e emerging role of the mito‐chondrial‐derived peptide humanin in stress resistance. J Mol Endo‐crinol 50:R11‐19.

Zhang SX, Sanders E, Fliesler SJ, Wang JJ (2014) Endoplasmic reticulum stress and the unfolded protein responses in retinal degeneration. Exp Eye Res 125:30‐40.

David R. Hinton, M.D., F.A.R.V.O., dhinton@usc.edu.

10.4103/1673-5374.198970

*< class="emphasis_italic">Correspondence to: David R. Hinton, M.D., F.A.R.V.O., dhinton@usc.edu.

orcid: 0000-0002-3971-8891 (David R. Hinton)