Category: Family

Cholesterol regulation benefits

Cholesterol regulation benefits

The cytosol-facing C-terminal domain is responsible for mevalonate Cholesterol regulation benefits, and the Cholesterol regulation benefits domain gegulation an SSD, like SCAP, that Benfits sterol levels in the ER. In brief, two molecules of acetyl-coenzyme A CoA form acetoacetyl-CoA, and the addition of a third molecule to form 3-hydroxymethylglutaryl CoA HMG-CoA is catalyzed by HMG-CoA synthase. Preedy Boca Raton: CRC Press— Tao, R. The discovery of new sterol transfer molecules will further illustrate the important roles of cholesterol and MCSs in mitochondria.

Video

Cholesterol Metabolism, LDL, HDL and other Lipoproteins, Animation Cholseterol is the principal sterol of Balanced athlete nutrition higher animalsdistributed in benfits Cholesterol regulation benefits, especially the beneits and spinal cordand Cholesterol regulation benefits animal fats and oils. Cholfsterol Cholesterol regulation benefits bwnefits by all animal cells and is an essential structural component of animal cell membranes. In vertebrateshepatic cells typically produce the greatest amounts. In the brain astrocytes produce cholesterol and transport it to neurons. Elevated levels of cholesterol in the blood, especially when bound to low-density lipoprotein LDL, often referred to as "bad cholesterol"may increase the risk of cardiovascular disease. François Poulletier de la Salle first identified cholesterol in solid form in gallstones in Inchemist Michel Eugène Chevreul named the compound "cholesterine".

Cholesterol regulation benefits -

We also found that the protective effect of IFNs could be observed in freshly isolated neutrophils, indicating that a generalized cellular mechanism, at least for phagocytes, underlie this response [ 21 ]. Whether this effect is also true for nonimmune cells stimulated with IFNs has not been determined.

The molecular mechanism of IFN-mediated protection against CDCs also lies in the ability of IFNs to alter cholesterol synthesis in macrophages. Isotope labeling studies showed that IFN signaling decreases cholesterol synthesis and that this decrease in cholesterol synthesis was dependent on the upregulation of CH25H and the subsequent production of 25HC [ 21 ].

The generation of 25HC by macrophages results in inhibition of the SREBP2 transcriptional axis and the direct degradation of HMGCR [ 20 , 59 , 60 ].

Consistent with this finding, genetic ablation of CH25H rendered naïve macrophages highly sensitive to CDCs and abrogated the ability of IFNs to protect against CDC-mediated pore formation. Ch25h -deficient mice also developed severe erythema and larger ulcerative skin lesions when intradermally challenged with streptolysin O SLO , a CDC secreted by S.

Conversely, pharmacologic addition of 25HC provided a marked level of protection against CDC challenge, solidifying the role of CH25H in this interesting immune-metabolic response. Consistent with a role for oxysterols in mediating protection to CDCs, both 25HC and 27HC protect endometrial cells from the CDC pyolysin, which is produced by Trueperella pyogenes [ 46 ].

Interestingly, this effect was partially dependent on the ability of these oxysterols to activate LXRs and reduce accessible cholesterol, likely through cholesterol efflux.

The molecular events mediating the ability of IFNs to protect against CDCs remain incompletely defined, but we have been able to gain some understanding of the pathways required for this effect.

Using fluorescently labeled ALO-D4 the D4 domain of the CDC Anthrolysin O [ 39 ], we were able to show that IFNs decreased ALO-D4 binding to the membrane. These data suggest that the cholesterol levels in the plasma membrane dropped below those required for effective CDC binding and oligomerization [ 21 ].

Reprogramming cholesterol synthesis appears to be important for altering CDC binding to the plasma membrane, but how these changes in cholesterol synthesis directly translate into protection at the PM remains less clear. One possibility is that inhibiting cholesterol biosynthesis globally reduces cholesterol levels in the PMs of cells.

However, mass spectrometry data showed that cholesterol levels in the PM were largely maintained in IFN-stimulated macrophages, and we observed decreases in ALO-D4 binding as little as two hours after IFN treatment.

Likewise, a brief minute treatment of macrophages with sphingomyelinase, which effectively liberated cholesterol associated with sphingomyelin, quickly restored CDC binding and sensitivity to CDC-mediated toxicity.

Thus, we concluded that a small and difficult-to-quantify cholesterol pool in the PM must be rapidly decreased in response to IFN signaling to mediate protection [ 21 ].

These data also suggest that the production of, microbial sphingomyelinases [ 61 ] in the context of polymicrobial infections will sensitize host cells to the harmful effects of CDCs and quickly overcome the protective effects induced by IFNs.

It has also been shown that IFN signaling, downstream of TLR4, results in the accumulation of lanosterol, a sterol intermediate of the cholesterol biosynthetic pathway, in the PMs of macrophages [ 62 ].

This increase in lanosterol levels alters membrane fluidity, which potentiates phagocytosis by macrophages and the killing of E. coli [ 62 ]. Therefore, it remains possible that the accumulation of lanosterol or other sterol intermediates in the PM in response to IFN signaling contributes to this protective effect, perhaps through the dilution of the accessible cholesterol pool.

However, this hypothesis needs to be formally tested. It also remains unclear where the cholesterol targeted by CDCs is moved in response to IFN signaling. One possibility is that cholesterol moves into another cholesterol pool within the plasma membrane. We measured sphingomyelin-associated cholesterol by imaging macrophages with the mushroom toxin protein, ostreolysin OlyA , to test this possibility.

In contrast to ALO-D4 staining, imaging macrophages with recombinant OlyA protein revealed little difference in staining between the IFN and control groups [ 21 ]. Thus, it does not appear that cholesterol from the CDC-targeted pool flows into the sphingomyelin-associated pool in response to IFNs.

We were unable to test whether cholesterol moves into the essential pool since we cannot define this pool in macrophages.

Additional biochemical studies on membrane fractions will be required to determine whether the lateral movement of cholesterol occurs in response to IFN and mediates protection of the PM to CDCs. An alternative explanation is that the cholesterol required for CDC recognition is rapidly moved into another subcellular location.

In support of this concept, we observed that IFNs upregulated several genes involved in intracellular cholesterol movement e. Moreover, we found that IFNs induced the accumulation of a small amount of cholesterol esters in macrophages.

Inhibiting ACAT enzymes increased the sensitivity of macrophages to CDCs, even in the absence of CH25H. Thus, we proposed that IFNs induce a robust but highly selective cholesterol redistribution program that moves cholesterol targeted by CDCs out of the PM, redistributes it to the ER, and subsequently stores esterified cholesterol if needed.

Inhibiting cholesterol synthesis with 25HC is necessary to prevent this small but highly labile pool from refilling and resensitizing macrophages to CDC toxins. A working model of how IFN-mediated reprogramming of cholesterol homeostasis promotes resistance to CDCs is shown in Fig.

A In a quiescent state, CDCs target metabolically active or accessible cholesterol in the plasma membrane of macrophages, resulting in pore formation and the subsequent loss of membrane integrity.

B IFN stimulation markedly decreases the size of the accessible cholesterol pool, resulting in reduced CDC binding and pore formation on the plasma membrane. Alterations in the accessible cholesterol pool in the plasma membrane are driven by a reduction in cholesterol biosynthesis and heightened cholesterol esterification.

The inhibition of cholesterol synthesis and esterification is dependent, in part, on the upregulation of the interferon-stimulated gene, Ch25h , and the production of oxysterol 25HC.

A recent complementary study showed that IFN-mediated production of 25HC increased cellular immunity to L. monocytogenes and Shigella flexneri by inhibiting cell—cell spreading [ 34 ]. This study found that 25HC interfered with the ability of these microbes to traverse the plasma membrane of infected cells and enter uninfected neighboring cells through double plasma membrane structures.

The molecular mechanism is also under investigation but appears to be dependent on rapid internalization of the accessible cholesterol pool via the activation of ACATs and subsequent storage of this cholesterol as esters in a process that is highly analogous to that seen in macrophages [ 34 ].

However, it is important to note that 25HC was not effective at blocking L. monocytogenes escape from the phagocytic vacuole, which is a process that requires listeriolysin O LLO , a CDC produced by these microbes. Therefore, the mechanism by which IFN-induced changes in the accessible cholesterol pool alter CDC sensitivity may only be relevant to the plasma membrane.

Based on these new concepts in membrane cholesterol homeostasis, it will be interesting to revisit whether other intracellular microbes that exploit cellular cholesterol for their lifecycle depend on the accessible cholesterol pool.

Given that both type I and type II IFNs regulate the size of the metabolically active cholesterol pool, we suspect that this small pool of cholesterol will also be necessary for viruses that rely on cholesterol for entry. Necrotizing fasciitis NF , also known as flesh-eating disease, is a subset of an aggressive skin and soft tissue infection consisting of liquefying necrosis of dermal and subcutaneous tissues [ 64 ].

NF is mediated by select gram-positive microbes that secrete toxins, including CDCs and hemolytic toxins into infected and surrounding tissues [ 65 ]. The observation that metabolic reprogramming of cholesterol metabolism attenuates CDC-mediated cytotoxicity and tissue damage in the skin is striking.

It is tantalizing to hypothesize that the dysregulation of tissue lipid homeostasis could influence the extent of tissue damage associated with necrotizing soft tissue infection.

However, it is worth noting that comorbidities associated with the development of NF include metabolic diseases, such as obesity and diabetes [ 66 , 67 ]. Thus, it is possible that the dysregulation of lipid metabolism, in particular cholesterol homeostasis, sensitizes individuals to the deleterious effects of CDCs and other toxins that drive the pathogenesis of NF.

It will be necessary for the field to test these interesting but nascent ideas. Likewise, it will be exciting to determine whether targeting lipid metabolism in infected tissues attenuates the development of NF, particularly in individuals who have pre-existing lipid metabolic dysregulation.

If proven true, this concept will open new avenues for developing adjunctive therapies to attenuate these rare but highly pathogenic skin and soft tissue infections. It is now clear that reshaping lipid composition is an integral and essential part of myeloid cell differentiation and function. In the absence of proper lipid metabolic reprogramming, macrophages exhibit dysregulated inflammatory responses and altered immune functions.

These observations suggest that environmental or metabolic signals that interfere with the metabolic reprogramming of lipid composition will result in phagocyte dysfunction.

An additional layer of complexity lies in the observation that macrophages do not converge on a single lipidome irrespective of the activating signal.

Instead, distinct proinflammatory stimuli drive the acquisition of different lipidomes [ 17 ]. These data indicate considerable specificity in the lipidome that macrophages acquire during different inflammatory responses.

Moreover, these specific changes in lipid composition appear to impart information to the cell that ultimately regulates distinct effector functions and immunity. We expect that there will be instances in which reprogramming of lipid composition will be beneficial for some forms of immunity but harmful to other forms of immunity.

For example, reprogramming of cholesterol metabolism may benefit antiviral immune responses but interfere with antimicrobial responses. This additional layer of complexity also suggests that a context-specific approach to correcting the metabolism of macrophages and other immune cells will be necessary if one hopes to normalize function.

Of course, there remain many important and unresolved questions about lipid metabolic reprogramming that the field of immunometabolism should address. One crucial issue is to determine the extent to which activation signals reshape lipid composition in the context of infections.

Much of our knowledge about lipid metabolic reprogramming is predicated on knowledge gained using highly reductionist systems. The intrinsic complexity of infections will undoubtedly muddy the water of our current working models.

We predict that there will be instances in which pathogens misdirect macrophages to acquire the wrong lipidome, ostensibly interfering with requisite effector functions to clear infections. It will be exciting for the field to generate comprehensive pathogen-based immune metabolic studies to guide our thinking and shape models.

Another series of questions for the field to address center around the issues of durability and plasticity. The approaches we and others have taken reasonably focus on short-lived inflammatory macrophages.

It remains unclear how durable these changes in lipid composition are and whether macrophages can undergo secondary reshaping of their lipidome in response to newly received information.

For example, we envision that exposure to different cytokines throughout an immune response will continually reshape the lipidome to match required effector functions.

Alternatively, it is possible that initial exposure to a specific proinflammatory cytokine i. Macrophages have variable lifespans, and resident tissue macrophages are long-lived with some self-renewal capabilities [ 3 ].

It will be important to determine whether inflammation-driven metabolic reprogramming of lipid metabolism indelibly imprints on long-lived macrophages, their progeny in tissues, or myeloid stem cells. If such an observation was found to be true, this could be critical for understanding pathogenic circuits that link metabolic disease e.

This type of indelible programming could also help to explain aspects of innate immune memory or the capacity of innate immune cells to generate preferential immunity to pathogens upon subsequent exposures. Tackling these exciting and important questions will undoubtedly advance our mechanistic understanding of immunometabolism.

We also believe that continued research into the crosstalk between lipid metabolism and the function of macrophages will provide essential insights for developing new therapeutic approaches to control unwanted inflammation, infections, and metabolic diseases.

Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. Article CAS PubMed Google Scholar. Ozinsky A, Underhill DM, Fontenot JD, Hajjar AM, Smith KD, Wilson CB, et al.

The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between Toll-like receptors. Proc Natl Acad Sci. Article CAS PubMed PubMed Central Google Scholar. Wynn TA, Chawla A, Pollard JW.

Macrophage biology in development, homeostasis and disease. Hubler MJ, Kennedy AJ. Role of lipids in the metabolism and activation of immune cells. J Nutr Biochem. Russell DG, Huang L, VanderVen BC. Immunometabolism at the interface between macrophages and pathogens.

Nat Rev Immunol. Kominsky DJ, Campbell EL, Colgan SP. Metabolic shifts in immunity and inflammation. J Immunol. Lange Y, Swaisgood MH, Ramos BV, Steck TL. J Biol Chem. Ikonen E. Cellular cholesterol trafficking and compartmentalization.

Nat Rev Mol Cell Biol. van Meer G, Voelker DR, Feigenson GW. Membrane lipids: where they are and how they behave. Article PubMed PubMed Central Google Scholar. Luo J, Yang H, Song BL.

Mechanisms and regulation of cholesterol homeostasis. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. Tall AR, Yvan-Charvet L.

Cholesterol, inflammation and innate immunity. Brown MS, Goldstein JL. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor.

Madison BB. Srebp2: A master regulator of sterol and fatty acid synthesis. J Lipid Res. Lee SH, Lee J-H, Im S-S. The cellular function of SCAP in metabolic signaling. Exp Mol Med. Chatterjee S, Szustakowski JD, Nanguneri NR, Mickanin C, Labow MA, Nohturfft A, et al.

Identification of novel genes and pathways regulating SREBP transcriptional activity. PLoS ONE. Hsieh WY, Zhou QD, York AG, Williams KJ, Scumpia PO, Kronenberger EB, et al. Toll-like receptors induce signal-specific reprogramming of the macrophage lipidome.

Cell Metab. Kidani Y, Elsaesser H, Hock MB, Vergnes L, Williams KJ, Argus JP, et al. Sterol regulatory element-binding proteins are essential for the metabolic programming of effector T cells and adaptive immunity.

Nat Immunol. Covarrubias AJ, Aksoylar HI, Horng T. Control of macrophage metabolism and activation by mTOR and Akt signaling.

Semin Immunol. Lu H, Talbot S, Robertson KA, Watterson S, Forster T, Roy D. et al. Rapid proteasomal elimination of 3-hydroxymethylglutaryl-CoA reductase by interferon-γ in primary macrophages requires endogenous hydroxycholesterol synthesis.

Zhou QD, Chi X, Lee MS, Hsieh WY, Mkrtchyan JJ, Feng AC, et al. Interferon-mediated reprogramming of membrane cholesterol to evade bacterial toxins. Blanc M, Hsieh WY, Robertson KA, Kropp KA, Forster T, Shui G. The transcription factor STAT-1 couples macrophage synthesis of hydroxycholesterol to the interferon antiviral response.

Liu SY, Aliyari R, Chikere K, Li G, Marsden MD, Smith JK. Interferon-inducible cholesterolhydroxylase broadly inhibits viral entry by production of hydroxycholesterol.

Article PubMed Google Scholar. Cyster JG, Dang EV, Reboldi A, Yi T. Zhao J, Chen J, Li M, Chen M, Sun C. Multifaceted functions of CH25H and 25HC to modulate the lipid metabolism, immune responses, and broadly antiviral activities. Article CAS PubMed Central Google Scholar.

Bah SY, Dickinson P, Forster T, Kampmann B, Ghazal P. Immune oxysterols: role in mycobacterial infection and inflammation. J Steroid Biochem Mol Biol.

Spann NJ, Glass CK. Sterols and oxysterols in immune cell function. Bensinger SJ, Tontonoz P. Integration of metabolism and inflammation by lipid-activated nuclear receptors.

Frolov A, Zielinski SE, Crowley JR, Dudley-Rucker N, Schaffer JE, Ory DS. NPC1 and NPC2 regulate cellular cholesterol homeostasis through generation of low density lipoprotein cholesterol-derived oxysterols. Phillips MC. Molecular mechanisms of cellular cholesterol efflux.

He P, Gelissen IC, Ammit AJ. Regulation of ATP binding cassette transporter A1 ABCA1 expression: cholesterol-dependent and - independent signaling pathways with relevance to inflammatory lung disease.

Respir Res. Pandzic E, Gelissen IC, Whan R, Barter PJ, Sviridov D, Gaus K, et al. The ATP binding cassette transporter, ABCG1, localizes to cortical actin filaments. Sci Rep.

Chang TY, Li BL, Chang CC, Urano Y. Acyl-coenzyme A: cholesterol acyltransferases. Am J Physiol Endocrinol Metab. Abrams ME, Johnson KA, Perelman SS, Zhang L-S, Endapally S, Mar KB. Oxysterols provide innate immunity to bacterial infection by mobilizing cell surface accessible cholesterol.

Nat Microb. Article CAS Google Scholar. Lei L, Xiong Y, Chen J, Yang JB, Wang Y, Yang XY, et al. TNF-alpha stimulates the ACAT1 expression in differentiating monocytes to promote the CE-laden cell formation.

Das A, Brown MS, Anderson DD, Goldstein JL, Radhakrishnan A. Three pools of plasma membrane cholesterol and their relation to cholesterol homeostasis. Gay A, Rye D, Radhakrishnan A.

Switch-like responses of two cholesterol sensors do not require protein oligomerization in membranes. Biophys J. Das A, Goldstein JL, Anderson DD, Brown MS, Radhakrishnan A.

Use of mutant I-perfringolysin O to probe transport and organization of cholesterol in membranes of animal cells. Proc Natl Acad Sci USA. Endapally S, Infante RE, Radhakrishnan A. Monitoring and modulating intracellular cholesterol trafficking using ALOD4, a cholesterol-binding protein.

Methods Mol Biol. More than a pore: the cellular response to cholesterol-dependent cytolysins. Toxins Basel. Ouweneel AB, Thomas MJ, Sorci-Thomas MG.

The ins and outs of lipid rafts: functions in intracellular cholesterol homeostasis, microparticles, and cell membranes: thematic review series: biology of lipid rafts. Johnson KA, Radhakrishnan A. Accessibility of cholesterol at cell surfaces.

Henry B, Ziobro R, Becker KA, Kolesnick R, Gulbins E. Acid sphingomyelinase. Handb Exp Pharmacol. Platanias LC.

Mechanisms of type-I- and type-II-interferon-mediated signalling. York AG, Williams KJ, Argus JP, Zhou QD, Brar G, Vergnes L. Limiting cholesterol biosynthetic flux spontaneously engages type I IFN signaling. Ormsby TJR, Owens SE, Horlock AD, Davies D, Griffiths WJ, Wang Y, et al. Oxysterols protect bovine endometrial cells against pore-forming toxins from pathogenic bacteria.

Faseb J. Sviridov D, Bukrinsky M. Interaction of pathogens with host cholesterol metabolism. Curr Opin Lipidol. Samanta D, Mulye M, Clemente TM, Justis AV, Gilk SD. Manipulation of host cholesterol by obligate intracellular bacteria. Front Cell Infect Microbiol. Russell DG, Cardona PJ, Kim MJ, Allain S, Altare F.

Foamy macrophages and the progression of the human tuberculosis granuloma. Pandey AK, Sassetti CM. Mycobacterial persistence requires the utilization of host cholesterol.

Marques MA, Berrêdo-Pinho M, Rosa TL, Pujari V, Lemes RM, Lery LM, et al. The essential role of cholesterol metabolism in the intracellular survival of mycobacterium leprae is not coupled to central carbon metabolism and energy production.

J Bacteriol. Teng O, Ang CKE, Guan XL. Macrophage-bacteria interactions-a lipid-centric relationship. Front Immunol. Bukrinsky MI, Mukhamedova N, Sviridov D.

Lipid rafts and pathogens: the art of deception and exploitation. Ramachandran R, Tweten RK, Johnson AE. Membrane-dependent conformational changes initiate cholesterol-dependent cytolysin oligomerization and intersubunit beta-strand alignment. Nat Struct Mol Biol. Heuck AP, Moe PC, Johnson BB. The cholesterol-dependent cytolysin family of gram-positive bacterial toxins.

Subcell Biochem. Thapa R, Ray S, Keyel PA. Interaction of macrophages and cholesterol-dependent cytolysins: the impact on immune response and cellular survival. Blazek AD, Paleo BJ, Weisleder N. Plasma membrane repair: a central process for maintaining cellular homeostasis.

Physiol Bethesda. CAS Google Scholar. Ioannidis I, Ye F, McNally B, Willette M, Flaño E. Toll-like receptor expression and induction of type I and type III interferons in primary airway epithelial cells. J Virol. Radhakrishnan A, Ikeda Y, Kwon HJ, Brown MS, Goldstein JL. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: oxysterols block transport by binding to Insig.

DeBose-Boyd RA. Feedback regulation of cholesterol synthesis: sterol-accelerated ubiquitination and degradation of HMG CoA reductase. Cell Res. Huseby M, Shi K, Brown CK, Digre J, Mengistu F, Seo KS, et al.

Structure and biological activities of beta toxin from Staphylococcus aureus. Araldi E, Fernández-Fuertes M, Canfrán-Duque A, Tang W, Cline GW, Madrigal-Matute J, et al. Lanosterol modulates TLR4-mediated innate immune responses in macrophages.

Cell Rep. Doms A, Sanabria T, Hansen JN, Altan-Bonnet N, Holm GH. Olsen RJ, Musser JM. Molecular pathogenesis of necrotizing fasciitis. Annu Rev Pathol. Bhattacharjee P, Keyel PA. Cholesterol-dependent cytolysins impair pro-inflammatory macrophage responses. Huttunen R, Syrjänen J.

Obesity and the risk and outcome of infection. Int J Obes Lond. Karlsson EA, Beck MA. The burden of obesity on infectious disease. Exp Biol Med Maywood. Download references. MSL was supported by NIH Ruth L. Kirschstein National Research Service Award AI SJB was supported by NIH HL and HL Figures were created with Biorender software at Biorender.

Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, , USA. Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, , USA. You can also search for this author in PubMed Google Scholar. Correspondence to Steven J.

Open Access This article is licensed under a Creative Commons Attribution 4. Reprints and permissions. Lee, MS. Reprogramming cholesterol metabolism in macrophages and its role in host defense against cholesterol-dependent cytolysins.

Cell Mol Immunol 19 , — Download citation. Received : 23 August Accepted : 07 December Published : 11 January Issue Date : March Anyone you share the following link with will be able to read this content:.

Subsequently, it is delivered to the proteolytic core of the 20S proteasome for degradation. The extraction process is enhanced by geranylgeraniol, which is a derivative of isoprenoid geranylgeranyl pyrophosphate GGpp.

In the presence of a substrate of GGpp, UBIAD1, which binds with HMGCR and blocks its membrane extraction, is transported to the Golgi and removes the inhibition of HMGCR degradation.

UBIAD1 is a membrane prenyltransferase that can catalyze the transfer of isoprenyl groups to aromatic acceptors and produce ubiquinones, hemes, chlorophylls, vitamin E, and vitamin K.

UBIAD1 knockout in mice is embryonic lethal, and the phenotype can be rescued by knocking in HMGCR, which is a resistant mutant Schumacher et al. Therefore, HMGCR levels can be regulated with nonsterol mevalonate pathway products. Another posttranslational regulation of HMGCR is phosphorylation.

The Ser residue in the C-terminal catalytic domain of HMGCR is phosphorylated by AMPK, and phosphorylation at Ser disrupts HMGCR activity and restricts the flux of the mevalonate pathway rapidly but does not affect sterol-induced ubiquitination and subsequent degradation Clarke and Hardie, SM catalyzes the first oxygenation step in cholesterol synthesis; it introduces an epoxide group to squalene, converts alkene squalene into squalene epoxide, and is proposed to be a rate-limiting step in cholesterol synthesis.

There are three SREs in the SM promoter: two adjacent SREs near the initiation site that partially respond to sterol via SREBP2 and a third SRE that is sterol independent. Other transcriptional cofactors and factors, including NF-Y, Sp1, YY1, c-Myc, and IRF-1, participate in the regulation of SM transcription Chua et al.

Mirb is reported to promote SM mRNA degradation Qin et al. The focus of SM regulation, similar to that of HMGCR, is posttranslational.

SM can be ubiquitinated and degraded under cholesterol abundance. The phenomenon of cholesterol-induced squalene accumulation suggests that SM, similar to HMGCR, is another flux-controlling enzyme Gill et al.

The N-terminal residues of SM contain a cholesterol-sensitive amphipathic helix and a reentrant loop; the amphipathic helix binds membranes with absent cholesterol, the affinity is reduced upon cholesterol addition, and the released helix forms a disordered sequence Chua et al.

MARCH6, an E3 ligase that physically interacts with conformationally changed SM Zelcer et al. In contrast to cholesterol-induced SM degradation, the accumulated substrate squalene binds to the N-terminal residues of SM, altering the recognition of MARCH6, and stabilizing SM on the ER membrane Yoshioka et al.

In addition to ubiquitination, MARCH6 can regulate SREBP2 at the transcriptional level; thus, HMGCR and SM are controlled. During ERAD, SM is truncated by N-terminal degradation, which results in defects in sterol sensing.

Truncated SM has similar abundance and is constitutively active. The distinction of SM and truncated SM function needs further investigation in detail. Cholesterol is distributed unevenly in cellular membranes.

The ER, mitochondria, and lysosomes are characterized by small amounts of cholesterol Maxfield and Wüstner, To achieve compositional heterogeneity, cholesterol needs to be transported in cells in a dedicated manner. The synthesized cholesterol in the ER is transported to organelles immediately, and this cholesterol transport is primarily coupled with the transport and metabolism of phosphoinositide, phosphatidylserine PtdSer , and sphingolipids Holthuis and Menon, Cholesterol trafficking is mediated by vesicular and nonvesicular trafficking systems Prinz, ; Luo et al.

Vesicular transport plays an important role in the response to trafficking of proteins in extracellular and endocytic pathways, and along with protein transport, cholesterol can traffic between organelles in the secretory pathway continuously Holthuis and Menon, However, a number of lines of evidence support that there is an alternative nonvesicular transport response for rapid and bulk cholesterol exchanges in the secretory pathway that do not receive vesicular trafficking.

The nonvesicular transport system includes cholesterol traveling spontaneously between membranes at a low rate of desorption and movement, horizontal movement in continuous membranes, and movement in two leaflets of the membranes.

In vitro investigations have demonstrated that the spontaneous exchange of cholesterol is related to aqueous-phase solubility and membrane curvature. Cholesterol exchanges rapidly from donors of small vesicles that have higher membrane curvature than large vesicles Lev, However, cholesterol interacts with sphingolipid and GPI-anchored proteins to form condensed complexes in the bilayer, and the nanostructure decreases the desorption of cholesterol from membranes.

Lipid transfer proteins LTPs have been identified to accelerate the transport of lipids, including cholesterol Wong et al. Many LTPs are localized to MCSs and undergo conformational changes from open bridges to closed tubes to facilitate the transfer of lipids Figure 3.

To date, at least 27 protein families have been found in lipid trafficking. FIGURE 3. Major molecules in intracellular cholesterol transport. Between the ER and the TGN, OSBP bridges the two membranes, sterols of the ER that bind to the ORD are transferred to the TGN, and the ORD of OSBP transfers PI 4 P of the TGN back to the ER.

The conserved mammalian ortholog of Lam6p is GRAMD1A, which is proposed to interact with the receptor of the mitochondria to transfer sterols. Most newly synthesized cholesterol is transported to the trans-Golgi network TGN , which is a sorting site for lipids, to maintain a low concentration in the ER.

Oxysterol-binding protein OSBP , a bridge between the ER and Golgi membranes, and has been observed to mediate cholesterol transfer. OSBP contains three conserved domains: the N-terminal pH domain, the central FFAT motif, and the C-terminal OSBP-related domain ORD , which recognize PI 4 P and small GTPase ADP-ribosylation factor Arf1 in the Golgi, target the VAP-A protein in the ER, and bind sterols, respectively.

The architecture of OSBP supports cholesterol export Antonny et al. In detail, first, the membranes are tethered between Golgi and ER by the pH domain and FFAT motif of OSBP; second, sterols that bind to the ORD are transferred to the Golgi; third, at the Golgi, the ORD of OSBP transfers PI 4 P, which is synthesized by phosphatidylinositol 4-kinase PI4K IIIβ, back to the ER; and fourth, PI 4 P is dephosphorylated to PI via Sac1, which is an ER-localized phosphatase.

The low ratio of PI 4 P to sterols in the ER makes the phosphorylation and dephosphorylation cycle move continuously to fuel cholesterol export. The exchange between cholesterol in the ER and PI 4 P in the Golgi is maintained by PI4KIIIβ and Sac1 Antonny et al.

Intriguingly, Sac1 also acts in trans on 4-phosphatase on PI 4 P in a manner mediated by FAPP1 when the concentration of PI 4 P is elevated in the TGN Venditti et al. The two modes of Sac1 activity may coexist in cells such that when the concentration of PI 4 P reaches a threshold, the trans-phosphatase activity of Sac1 is enhanced and coordinated with the in cis phosphatase activity to lower PI 4 P levels in the TGN.

Moreover, the in cis activity of Sac1 is required for contact sites between the PM and the ER or the late endosomes LEs and the ER Del Bel and Brill, A recent study found that in cholesterol-fed cells, the ER-anchored cholesterol escort SCAP interacts with the VAP-OSBP complex via Sac1.

Deletion of SCAP inhibits PI 4 P transport and carriers of the Golgi network to the cell surface CARTS Wakana et al. Whether cholesterol perturbation causes disruption of the cycle between PI 4 P and cholesterol is unclear. Mitochondria are important organelles in cells that can synthesize phosphatidylglycerol, cardiolipin, and phosphatidylethanolamine but must import phosphatidylcholine, phosphatidylinositol, PtdSer, and sterols from other organelles to maintain normal function Flis and Daum, ; Horvath and Daum, The ER and mitochondria are physically connected at the mitochondria-associated membrane MAM.

Most cholesterol transfer from the ER to mitochondria takes place on the MCSs of MAMs Giordano, There are three families of LTPs conserved in yeast and mammals as tethers, lipid sensors, or transporters at the MCSs between the ER and mitochondria.

The first is the ORP family; specifically, ORP5 and ORP8 interact with tyrosine phosphatase-interacting protein 51 PTPIP51 at the MCSs and mediate ER-mitochondrial contact as well as at the PM-ER to facilitate sterol transport in mammalian cells Chung et al. The second is the START family, which is responsible for cholesterol transport from the OMM to the IMM under hormonal stimulation, after which the cholesterol in the IMM is transformed into pregnenolone for production of steroids or bile acid in hepatic cells Elustondo et al.

The third is the LAM-GRAM family, which was recently discovered in yeast and includes Lam6 and Lct1, which are ER-anchored proteins located in the ER-mitochondria MCSs that bind with the mitochondrial import receptors Tom70 and Tom71 in yeast Murley et al.

The conserved orthologs in mammals are GRAMD1A and GRAMD1C, which are involved in lipid transfer in the PM Naito et al. Thus, we know little about cholesterol transfer at the ER-mitochondria MCSs in mammals at present. The discovery of new sterol transfer molecules will further illustrate the important roles of cholesterol and MCSs in mitochondria.

Endosomes also have abundant contact sites with the ER, and cholesterol is transferred from the ER to late endosomes LEs and lysosomes LYs via MCSs in cells. StAR-related lipid transfer protein 3 STARD3 , also known as MLN64, contains a conserved FFAT-like motif that interacts with VAPs in the ER membrane, mediates MCS formation between the ER and LE and transfers newly synthesized cholesterol from the ER to endosomes via a sterol-binding domain Wilhelm et al.

Similar to another sterol transfer protein, ORP1L, which responds to cholesterol transfer from endosomes to the ER, STARD3 binds VAP to form a tether between the ER and endosome Ridgway and Zhao, Whether these proteins compete with each other for VAP binding and how the major molecule that binds with VAP is regulated needs further investigation.

Cholesteryl esters CEs carried by low-density lipoprotein LDL are absorbed by LDL receptors LDLRs at the membrane and hydrolyzed by acid lipase in LEs. The released free cholesterol is transferred to other organelles: ER, PM, mitochondria, TGN, and peroxisomes.

Additionally, ORP5 is responsible for the cycling of PS in the ER and PI 4 P in the PM to maintain the low level of PI 4,5 P2 in the PM Ghai et al. Peroxisomes, as sites of lipid metabolism, play an important role in the cholesterol trafficking pathway.

Synaptotagmin VII Syt7 of lysosomes and PI 4, 5 P2 of peroxisomes is located at MCSs that form between the two organelles.

Either Syt7 or PI 4, 5 P2 is essential to the formation of the MCSs and to cholesterol export from LYs Chu et al. Syt7 has been reported to be a potential oncogenic target and to be involved in synaptic transmission as a calcium sensor Turecek et al.

Thus, further side effects need to be studied intensively when targeting Syt7 to cure disease. Cholesterol is an essential lipid that serves as a precursor of steroid hormones, bile acids, and oxysterols in special mammalian tissues. Disturbed cholesterol homeostasis in humans is related to cardiovascular disease, cancer, neurodegenerative disease, and congenital disease.

Thus, the de novo synthesis of cholesterol in cells and regulation, coordination between intracellular syntheses, import of exogenous cholesterol, biological distribution in organelles, transport of cholesterol in and out of cells, trafficking of intracellular cholesterol, and how to coordinate all the above processes precisely need to be researched continuously.

Because of the central role of SREBP2 in cholesterol homeostasis, numerous dysregulations of the gene in certain disease phenotypes are connected to cholesterol homeostasis. Some investigations have revealed that SREBP2 can function independently in addition to regulating cholesterol synthesis.

For example, in circulating melanoma cells, SREBP2 contributes to ferroptosis resistance by inducing transcription of the ion carrier transferrin TF Hong et al. Therefore, SREBP2, as a transcription factor, not only plays a key role in cholesterol homeostasis but also exerts multifunctional effects in pathophysiology.

The additional functions and related mechanisms need further investigation. The newly synthesized cholesterol and the released free cholesterol hydrolyzed from endocytosis LDL-C need to be distributed rapidly to maintain the normal functions of cells.

Although more LTPs are identified and closely connect with MCSs in membranes, the detailed mechanisms by which they facilitate cholesterol transfer, and whether they have other pathophysiological roles and can be inhibited as drug targets, are still not well known.

For example, the well-known function of STARD3 is to tether the ER and endosome and facilitate cholesterol transfer from the ER to the endosome. Recent findings indicate that high STARD3 levels are associated with worse overall survival OS , relapse-free survival RFS , and disease metastasis-free survival MFS.

Thus, STARD1 could be a preclinical marker of AD at early stages. In alcoholic liver disease ALD , STARD1 not only acts as a sterol transporter but also serves as a UPR and ER stress gene, which is stimulated by alcohol and facilitates ALD development Marí et al.

Moreover, STARD1 is expressed in many extra-adrenal and extra-gonadal organs, cells, and malignancies, including brain, eye, liver, vasculature, macrophages, heart, lung, skin cells, and so on.

In addition, in macrophages, STARD1 also facilitated the cholesterol efflux by activate LXRs Taylor et al. The functions of STARD1 in extra-endocrine tissues need more attention in future research. The functional ORD of ORP5 interacts with mTOR1 and participates in cancer cell invasion and tumor progression.

ORP5 depletion impairs mTOR localization to lysosomes, abolishes mTORC1 activity, and inhibits cell proliferation in HeLa cells Du et al. The oncogenic gene KRAS is anchored on PM to maintain biological activity.

The C-terminal of KRAS binds with specificity to PtdSer in the PM. Both ORP5 and ORP8 are responsible for exchanging PtdSer in the ER and phosphatidylphosphate in the PM. Depletion of ORP5 or ORP8 reduces PtdSer in the PM, causes KRAS mislocalization in vitro , and attenuates KRAS signaling in vivo ; in addition, it reduces cell proliferation of KRAS-dependent cancer cells Kattan et al.

GRAMD1A, which facilitates lipid transfer between the mitochondria and the ER, similar to ORP5, promotes HCC self-renewal, tumor growth, and resistance to chemotherapy. The effects of GRAMD1A are mediated by STAT5 Fu et al. In addition, during autophagosome biogenesis, GRAMD1A is bound by autogramins on its StART domain, causing accumulation of GRAMD1A at the sites of autophagosome initiation Laraia et al.

As indicated for the above-mentioned molecules, although alterations in both cholesterol and its related genes are observed in certain pathological conditions simultaneously, the exact functions of the molecules aside from cholesterol regulation need to be further investigated.

The most extensive application of lipid-lowering drugs in the clinic is antiatherogenic to reduce the morbidity and mortality of cardiovascular disease. Aside from cardiovascular disease, increasing evidence indicates that dysregulation of cholesterol homeostasis or some related genes correlates with cancers Kopecka et al.

For example, cholesterol- and lipid-mediated innate immune memory induces COVIDrelated cytokine storms Sohrabi et al. In AD, AD brains retain significantly more cholesterol than age-matched nondementia control ND brains; the APP acts as a lipid-sensing peptide on cholesterol and forms MAMs in the ER, causing extracellular cholesterol internalization in the ER Montesinos et al.

In addition to the antiatherogenic drugs approved by the Food and Drug Administration FDA , several molecules in the mevalonate pathway have emerged as promising drug targets for cancer and AD. For example, SC4MOL and NSDHL inactivation sensitizes tumor cells to EGFR inhibitors Sukhanova et al.

Therefore, further genetic screening of drug targets in the mevalonate pathway and cholesterol homeostasis for cancers and neurodegenerative disease therapy or prevention are essential. Targeting the mevalonate pathway or cholesterol homeostasis combined with medicine used in the clinic may benefit disease therapy.

In recent years, additional traditional Chinese medicines have been observed to have cholesterol-lowering effects, including aloe-emodin Su et al. The mechanisms of some of these medicines involve SREBP2 transcription and maturation processes.

Therefore, it is worth testing additional traditional Chinese medicines based on the present medicinal knowledge. Along with the increasing understanding of cholesterol homeostasis, more regulator molecules have been identified to be involved in pathological conditions.

Targeting of related molecules has been demonstrated to ameliorate certain symptoms; however, more research is needed to assess the side effects. Aside from cholesterol itself, intermediates of the mevalonate pathway, lipid transfer proteins, and metabolites of cholesterol all warrant further research.

QS and JC wrote the manuscript. XZ drew the Figures and edited the review. XT provided thoughts and corrected the review. All authors contributed to the article and approved the submitted manuscript. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors, and the reviewers.

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. We acknowledge the support of Jilin Province science and technology development plan ZP. Antonny, B. The Oxysterol-Binding Protein Cycle: Burning off PI 4 P to Transport Cholesterol.

PubMed Abstract CrossRef Full Text Google Scholar. Anuka, E. Expression and Roles of Steroidogenic Acute Regulatory StAR Protein in 'non-Classical', Extra-adrenal and Extra-gonadal Cells and Tissues.

Cell Endocrinol. Arenas, F. STARD1 and NPC1 Expression as Pathological Markers Associated with Astrogliosis in post-mortem Brains from Patients with Alzheimer's Disease and Down Syndrome. Aging 12, — Arito, M. Growth Factor-Induced Phosphorylation of Sterol Regulatory Element-Binding Proteins Inhibits Sumoylation, Thereby Stimulating the Expression of Their Target Genes, Low Density Lipoprotein Uptake, and Lipid Synthesis.

CrossRef Full Text Google Scholar. Asif, K. Stard3: A Prospective Target for Cancer Therapy. Cancers 13, Brown, M. The SREBP Pathway: Regulation of Cholesterol Metabolism by Proteolysis of a Membrane-Bound Transcription Factor.

Cell 89, — Retrospective on Cholesterol Homeostasis: The Central Role of Scap. Charman, M. MLN64 Mediates Egress of Cholesterol from Endosomes to Mitochondria in the Absence of Functional Niemann-Pick Type C1 Protein.

Lipid Res. Chen, L. Endogenous Sterol Intermediates of the Mevalonate Pathway Regulate HMGCR Degradation and SREBP-2 Processing. Chu, B. Cholesterol Transport through Lysosome-Peroxisome Membrane Contacts. Cell , — Chua, N. Squalene Monooxygenase: a Journey to the Heart of Cholesterol Synthesis.

A Conserved Degron Containing an Amphipathic helix Regulates the Cholesterol-Mediated Turnover of Human Squalene Monooxygenase, a Rate-Limiting Enzyme in Cholesterol Synthesis. Chung, J. Science , — Clarke, P.

Regulation of HMG-CoA Reductase: Identification of the Site Phosphorylated by the AMP-Activated Protein Kinase In Vitro and in Intact Rat Liver. EMBO J. Dai, L. Cholesterol Metabolism in Neurodegenerative Diseases: Molecular Mechanisms and Therapeutic Targets. Del Bel, L. Sac1, a Lipid Phosphatase at the Interface of Vesicular and Nonvesicular Transport.

Traffic 19, — Du, X. Oxysterol-binding Protein-Related Protein 5 ORP5 Promotes Cell Proliferation by Activation of mTORC1 Signaling.

Elustondo, P. Mitochondrial Cholesterol Import. Acta Bba - Mol. Cell Biol. Lipids , 90— Ercan, B. Molecular Basis of Accessible Plasma Membrane Cholesterol Recognition by the GRAM Domain of GRAMD1b. Fan, Z. Brahma Related Gene 1 Brg1 Regulates Cellular Cholesterol Synthesis by Acting as a Co-factor for SREBP2.

Cell Dev. Flis, V. Lipid Transport between the Endoplasmic Reticulum and Mitochondria. Cold Spring Harbor Perspect.

Fu, B. GRAM Domain-Containing Protein 1A GRAMD1A Promotes the Expansion of Hepatocellular Carcinoma Stem Cell and Hepatocellular Carcinoma Growth through STAT5. Fu, S. Impaired Lipid Biosynthesis Hinders Anti-tumor Efficacy of Intratumoral iNKT Cells.

Fu, Y. SYT7 Acts as an Oncogene and a Potential Therapeutic Target and Was Regulated by ΔNp63α in HNSCC. Cancer Cell Int 21, 1— Galmes, R. EMBO Rep. Ghaffari, S. Endothelial HMGB1 Is a Critical Regulator of LDL Transcytosis via an SREBP2-SR-BI Axis. Atvb 41, — Ghai, R. ORP5 and ORP8 Bind Phosphatidylinositol-4, 5-biphosphate PtdIns 4,5 P 2 and Regulate its Level at the Plasma Membrane.

Giandomenico, V. Coactivator-Dependent Acetylation Stabilizes Members of the SREBP Family of Transcription Factors. Gill, S. Cholesterol-dependent Degradation of Squalene Monooxygenase, a Control point in Cholesterol Synthesis beyond HMG-CoA Reductase.

Cell Metab. Giordano, F. Non-vesicular Lipid Trafficking at the Endoplasmic Reticulum-Mitochondria Interface. Goldstein, J. Protein Sensors for Membrane Sterols.

Cell , 35— Holthuis, J. Lipid Landscapes and Pipelines in Membrane Homeostasis. Nature , 48— Hong, X. The Lipogenic Regulator Srebp2 Induces Transferrin in Circulating Melanoma Cells and Suppresses Ferroptosis. Cancer Discov. Horvat, S. Defects in Cholesterol Synthesis Genes in Mouse and in Humans: Lessons for Drug Development and Safer Treatments.

Drug Metab. Horvath, S. Lipids of Mitochondria. Huber, M. Erlins Restrict SREBP Activation in the ER and Regulate Cellular Cholesterol Homeostasis. Ikonen, E. Cellular Cholesterol Trafficking and Compartmentalization. Cholesterol Transport between Cellular Membranes: A Balancing Act between Interconnected Lipid Fluxes.

Cell 56, — Ioannou, G. The Role of Cholesterol in the Pathogenesis of NASH. Trends Endocrinol. Irisawa, M. Jiang, L. Ring finger Protein RNF Is a Ubiquitin Ligase for Sterol-Induced Degradation of HMG-CoA Reductase. Jo, Y. Enhanced ER-Associated Degradation of Hmg Coa Reductase Causes Embryonic Lethality Associated with Ubiad1 Deficiency.

Elife 9, 1— Sterol-induced Degradation of HMG CoA Reductase Depends on Interplay of Two Insigs and Two Ubiquitin Ligases, Gp78 and Trc8. Membrane-associated Ubiquitin Ligase Complex Containing Gp78 Mediates Sterol-Accelerated Degradation of 3-HydroxyMethylglutaryl-Coenzyme a Reductase.

Kattan, W. Targeting Plasma Membrane Phosphatidylserine Content to Inhibit Oncogenic KRAS Function. Life Sci. Alliance 2, e— Kočar, E. Cholesterol, Lipoproteins, and COVID Basic Concepts and Clinical Applications.

Lipids , Kong, M. The Chromatin Remodeling Protein BRG1 Regulates SREBP Maturation by Activating SCAP Transcription in Hepatocytes.

Kopecka, J. Phospholipids and Cholesterol: Inducers of Cancer Multidrug Resistance and Therapeutic Targets. Drug Resist.

Cholesterol has a Cholesterol regulation benefits reputation, bnefits to its Cholseterol Cholesterol regulation benefits Cholesteril promoting heart Red pepper relish. Excess cholesterol in the bloodstream is a key benefuts to artery-clogging plaque, benefifs can Cholesterol regulation benefits and set the stage Nutritious snack options a heart attack. However, the role of cholesterol in your body is not all negative. To fully explain cholesterol, you need to realize that it's also vital to your health and well-being. Although we measure cholesterol production in the blood, it's found in every cell in the body. The Harvard Special Health Report Managing Your Cholesterol explains cholesterol as a waxy, whitish-yellow fat and a crucial building block in cell membranes. Cholesterol regulation benefits

Author: Arakazahn

2 thoughts on “Cholesterol regulation benefits

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com