Pericyte Loss in Diseases: Comparison
Please note this is a comparison between Version 2 by Peter Tang and Version 1 by Hongkuan Fan.

Pericytes are specialized cells located in close proximity to endothelial cells within the microvasculature. They play a crucial role in regulating blood flow, stabilizing vessel walls, and maintaining the integrity of the blood–brain barrier. The loss of pericytes has been associated with the development and progression of various diseases, such as diabetes, Alzheimer’s disease, sepsis, stroke, and traumatic brain injury. 

  • perictyes
  • vascular leak
  • diabetes
  • blood–brain barrier

1. Pericyte Loss in Diabetes

Diabetes is a chronic health condition that causes multiple vascular complications, such as retinopathy and nephropathy [23][1]. Pericyte loss is an early hallmark of diabetes-associated microvascular diseases and plays a crucial role in the disease progression of various organs including the retina, kidney, brain, and heart [23,24][1][2].

1.1. Pericyte Loss in Diabetic Retinopathy (DR)

DR, a major complication of diabetes, is the leading cause of blindness worldwide and is characterized by vascular damage in the retina [25,26,27][3][4][5]. Among the vascular cells, pericytes are the earliest to be affected by diabetes, and their loss is a hallmark of diabetic retinopathy, contributing to blood vessel leakage [28][6]. The loss of pericytes has been detected in the retinas of diabetic patients [29,30[7][8][9],31], as well as in various animal models, including mice [31[9][10][11],32,33], dogs [34,35][12][13], hamsters [36][14], and rats [37,38][15][16].
The precise mechanisms underlying pericyte loss in diabetic retinopathy are not yet fully understood. One hypothesis suggests that pericyte death via apoptosis is involved, as confirmed by studies in both patients and animal models of DR [30,32,39,40,41,42][8][10][17][18][19][20]. Mechanically, factors such as high-glucose [40,43][18][21], oxidative stress [44][22], advanced glycation end products [45[23][24],46], TNF-α [47][25], and IL-1β [48][26] have been shown to induce apoptosis in retinal pericytes in vitro and/or in vivo. Additionally, the migration of retinal pericytes may contribute to pericyte loss in DR, regulated by the Ang-Tie system and autophagy processes [49,50][27][28]. Pericyte loss contributes to EC-pericyte dissociation and vascular dysfunction as retinal capillary pericytes are critical to maintaining EC-pericyte contacts and the integrity of vascular barrier function via secretion of sphingosine 1-phosphate [51,52][29][30]. Therefore, preventing the loss of retinal pericytes would be beneficial. Pericytes derived from adipose-derived stem cells (ASCs) showed protective effects against capillary loss in the retina in a murine model of DR [53][31].

1.2. Pericyte Loss in Diabetic Nephropathy (DN)

As a common complication of diabetes, DN is characterized by proteinuria, microvascular damage, and the disruption of glomeruli and the tubular system [4,23,55,56][1][32][33][34]. It significantly impacts the quality of life of diabetic patients and is the leading cause of end-stage renal disease [23,56][1][34]. Renal pericyte-like cells, including peritubular pericytes, mesangial cells, and podocytes, are susceptible to oxidative stress induced by high glucose and play a critical role in the progression of DN [4,23][1][32].
Several hallmark features of DN, such as peritubular capillary rarefaction (PTC) and peritubular fibrosis, mesangial and glomerular hypertrophy, and podocyte injury, are closely associated with renal dysfunction [4,55,57,58,59][32][33][35][36][37]. Peritubular pericytes are crucial for maintaining the integrity of peritubular capillaries, as the loss of pericytes can accelerate PTC rarefaction [58,60,61][36][38][39]. Furthermore, the migration of peritubular pericytes away from the capillaries and their transformation into myofibroblasts are potential mechanisms underlying PTC and peritubular fibrosis [4,62,63][32][40][41]. However, the precise role and underlying mechanisms of peritubular pericytes in DN remain largely unknown. Mesangial cells, comprising approximately 30% of glomerular cells, undergo hypertrophy in the early stages of DN [4,23,64,65][1][32][42][43]. Increased mTOR activity may contribute to mesangial cell hypertrophy under high glucose conditions [4,65][32][43]. Moreover, diabetic rats and mice exhibit glomerular cell loss and apoptosis, which are associated with albuminuria and renal dysfunction [66,67][44][45]. Mechanistically, elevated levels of urinary miR-15b-5p have been observed in diabetic patients and db/db mice and contribute to high glucose-induced mesangial cell apoptosis [68][46]. Additionally, serum levels of Angpt2 were increased in diabetic patients and db/db mice, and the Angpt2/miR-33-5p/SOCS5 signaling pathway has been implicated in mesangial cell apoptosis under high glucose conditions [69][47].

1.3. Brain Pericyte Loss in Diabetes

Diabetes can induce damage that leads to dysfunction of the BBB and cognitive decline in both patients and experimental models [87,88,89,90][48][49][50][51]. Furthermore, diabetic patients have a higher risk of developing dementia-related diseases such as stroke and Alzheimer’s disease (AD) [91,92][52][53]. In the brain, diabetes-related complications are characterized by pericyte loss, increased BBB permeability, and neuronal dysfunction [87,89,93][48][50][54]. Reduced numbers of brain pericytes have been reported in diabetic patients [23[1][55],94], as well as in animal models of diabetes, including mice and rats [95,96,97][56][57][58]. In vitro studies have shown that oxidative stress induced by high glucose can lead to apoptosis in cultured brain pericytes [98,99,100,101][59][60][61][62]. The activity of mitochondrial carbonic anhydrases was believed to induce brain pericyte loss in diabetic mice as the inhibition of mitochondrial carbonic anhydrases activity can reduce oxidative stress and prevent pericyte dropout [96][57]. However, the exact mechanisms and in vivo processes underlying brain pericyte loss in diabetes require further investigation.

1.4. Cardiac Pericyte Loss in Diabetes

Cardiovascular disease (CVD) is a significant complication of diabetes and is the leading cause of heart failure or mortality in diabetic patients [102,103,104,105][63][64][65][66]. Pericytes play a crucial role, particularly in the early stages of diabetes-associated CVD, including myocardial and interstitial fibrosis [105,106,107][66][67][68]. The loss of pericytes has been demonstrated in the hearts of diabetic patients and diabetic pigs [108][69]. Additionally, studies by Tu et al. showed a reduction in the number of cardiac pericytes and microvascular coverage in diabetic mice [24][2]. The overexpression of thymosin beta 4 has the ability to mitigate cardiac pericyte loss in diabetic pigs, providing a potential therapeutic approach for diabetes-associated CVD [108][69]. However, the specific underlying mechanisms of cardiac pericyte loss in diabetes remain unclear and require further investigation.
In summary, pericyte loss is closely associated with various complications of diabetes and significantly contributes to disease development. Further research is needed to gain a better understanding of the underlying mechanisms involved and to explore novel therapeutic strategies targeting pericytes.

2. Pericyte Loss in Aging and Neurodegenerative Diseases

2.1. Pericyte Loss in Alzheimer’s Disease

AD is the most prevalent neurodegenerative disorder characterized by cognitive impairment, an accumulation of amyloid β-peptide (Aβ), BBB dysfunction, and neuroinflammation [109,110,111][70][71][72]. Pericytes play a critical role in AD, as their deficiency in mouse models of AD accelerates BBB breakdown and increases Aβ accumulation in the brain [112][73]. Pericyte loss has been reported in various regions of AD patients’ brains, including the white matter [113[74][75],114], precuneus [115][76], cortex [112[73][77],116], hippocampus [7[77][78],116], and retina [117][79]. Similarly, reduced pericyte numbers have been observed in the cortex [118[80][81],119], hippocampus [7[78][81],119], and retina [120][82] of AD mice. In the retina, the activation of inflammation appears to contribute to pericyte loss as an association between NF-κB p65 phosphorylation levels and vascular PDGFRβ expression was observed in AD mice [120][82]. Apoptosis is believed to contribute to pericyte loss, as pericyte apoptosis has been identified in the retina and hippocampus of AD patients [7,117][78][79]. In vitro studies have shown that Aβ stimulation induces apoptosis in cultured brain pericytes [7,119][78][81]. Mechanistically, decreased miR-181a levels and enhanced Fli-1 expression may contribute to pericyte loss and apoptosis in AD [7,119][78][81]. A reduced miR-181a expression has been observed in AD mice, but the overexpression of miR-181a can mitigate pericyte loss, improve BBB function, and decrease Aβ accumulation [119][81]. Furthermore, miR-181a inhibits Aβ-induced pericyte apoptosis in murine brain cell cultures [119][81].

2.2. Pericyte Loss in Amyotrophic Lateral Sclerosis (ALS)

ALS, a fatal neurodegenerative disorder, is characterized by blood–spinal cord barrier dysfunction and the progressive degeneration of motor neurons [125,126,127,128][83][84][85][86]. Recent studies have highlighted the important role of pericytes in ALS [129][87]. Decreased pericyte coverage or number has been observed in the ventral horn and spinal cord of ALS patients, which correlates with vascular disruption [130,131][88][89]. Furthermore, a loss of pericytes in the choroid plexus has been detected in patients with ALS, coupled with a deregulation of the blood–cerebrospinal fluid (CSF) barrier [132][90]. In a murine model of ALS, reduced pericyte coverage in spinal cord capillaries has also been demonstrated [133][91]. Interestingly, the administration of adipose-derived pericytes has shown promising results in ALS mice, extending their survival and increasing antioxidant enzymes in the brain [134][92]. These findings suggest that pericytes may represent a novel potential cell therapy for treating ALS, although further studies are needed to fully understand pericyte loss in ALS and its implications for disease progression.
Overall, pericyte loss in aging and neurodegenerative diseases poses a significant challenge that can have negative effects on brain health. Advancing the understanding of the underlying mechanisms of pericyte loss and developing new treatments to prevent or reverse this process are important areas of future research.

3. Pericyte Loss in Infectious Diseases

3.1. Pericyte Loss in Sepsis

Sepsis is a life-threatening condition caused by a microbial infection resulting in organ dysfunction and failure. It is characterized by a systemic inflammatory response and microvascular dysfunction [135,136][93][94]. Recent studies have highlighted the role of dysfunctional pericytes in sepsis-induced microvascular dysfunction, which serves as a hallmark of severe sepsis and septic shock [15,137][95][96]. Research by Nishioku et al. demonstrated the detachment of pericytes from the basal lamina in the hippocampus of LPS-treated mice [138][97]. The detachment of pericytes may contribute to sepsis-induced BBB dysfunction [139][98] as pericytes control vascular permeability in the brain [140][99]. Pericyte loss has also been observed in the lungs and hearts of LPS-treated mice, although this loss is not caused by apoptosis [141][100]. Reduced pericyte coverage in mesenteric microvessels has been demonstrated in both cecal ligation and puncture (CLP) and LPS-induced septic rats [142][101].

3.2. Pericyte Loss in HIV

The neurocognitive disorder is a major complication of HIV as the virus enters the brain shortly after infection, leading to inflammation and BBB disruption [146,147][102][103]. In vitro studies have demonstrated that cultured brain pericytes can be infected by HIV, resulting in enhanced production of inflammatory mediators and disruption of endothelial barrier properties [148,149][104][105]. Furthermore, evidence from in vivo studies, including HIV patients and mouse models of HIV, has shown that brain pericytes can be infected by HIV [150,151,152][106][107][108]. Following HIV infection, a reduction in pericyte coverage has been observed in the brains of HIV patients [150,153,154][106][109][110]. Similar pericyte loss has also been detected in the brains of mouse models of HIV and SIV-infected macaques [153,154][109][110]. It has been suggested that the higher concentration of PDGF-BB induced by HIV Tat via the activation of mitogen-activated protein kinases and nuclear factor-κB pathways may drive HIV-induced pericyte loss in the brain [153,155][109][111]. However, the role of pericytes in HIV has not been extensively examined. A better understanding of pericyte dysfunction and loss in the context of HIV may provide opportunities for the development of novel therapeutics.

3.3. Pericyte Loss in COVID-19

COVID-19 is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and affects various organs, including the heart, brain, and lungs [156,157,158,159][112][113][114][115]. Cardiac pericytes, which express high levels of angiotensin-converting enzyme 2 (ACE-2), the main receptor for SARS-CoV-2, are major targets for viral infection [160,161,162][116][117][118]. The infection of pericytes via SARS-CoV-2 contributes to cardiac complications associated with COVID-19, such as thrombosis, inflammation, and hemodynamic disturbances [163][119]. Studies have shown a significant loss of pericyte coverage in the heart capillaries of hamsters infected with SARS-CoV-2 [164][120]. Additionally, SARS-CoV-2 can infect cardiac pericytes, and its spike protein may induce pericyte dysfunction via CD147 receptor-mediated signaling pathway, leading to microvascular injury [157][113]. Brain pericytes, which also express ACE-2, are susceptible to SARS-CoV-2 infection, potentially driving inflammation and vascular dysfunction [158,165,166][114][121][122]. Patients with COVID-19 have shown lower levels of the pericyte marker PDGFRβ in their cerebrospinal fluid [158][114]. SARS-CoV-2 spike protein has been found to deregulate vascular and immune functions in brain pericytes [167][123], while the SARS-CoV-2 envelope protein has been shown to induce brain pericyte death in vitro [168][124]. In the lung, pericytes were infected by SARS-CoV-2 and are detached from pulmonary capillary endothelium in COVID-19 patients [159,169][115][125]. However, the underlying mechanisms of pericyte loss in COVID-19 remain largely unknown, and further studies are needed to investigate the role of pericytes, particularly in long COVID-19 [170][126].
Overall, the loss of pericytes in infectious diseases can have significant negative effects on patient outcomes. Understanding the mechanisms underlying pericyte loss in these diseases is crucial for the development of new treatments that can prevent or reverse this process and improve patient outcomes. Further research is needed to gain a better understanding of the role of pericytes in infectious diseases and to explore novel therapeutic approaches that can target these cells.

4. Pericyte Loss in Brain Injury

4.1. Pericyte Loss in Stroke

Stroke, a leading cause of death and disability worldwide, is associated with pericyte dysfunction and BBB disruption [171][127]. Pericytes play a critical role in regulating inflammation, angiogenesis and BBB function during stroke [171,172][127][128]. A rapid reduction in brain pericyte number and coverage has been observed in human stroke cases as well as in experimental stroke models, including mice and rats, following ischemic damage [113,173,174,175,176][74][129][130][131][132]. Pericyte apoptosis and autophagy have been detected in the brain from murine models of stroke, which may contribute to pericyte loss and BBB disruption [174,175][130][131]. The loss of regulator of G protein signaling 5 (RGS5) has been associated with increased pericyte number and improved BBB function in a mouse model of stroke, suggesting a role for RGS5 in brain pericyte loss during stroke [177][133]. Additionally, the inhibition of Sema3E/PlexinD1 signaling has been shown to increase pericyte number and enhance blood–brain barrier integrity in aged rats with stroke, further implicating this signaling pathway in brain pericyte loss [176][132]. Moreover, the deletion of hypoxia-inducible factors (HIF)-1 in pericytes has been found to prevent brain pericyte apoptosis and reduce vascular permeability in mice with stroke, indicating the involvement of pericyte HIF-1 in stroke-induced pericyte apoptosis [175][131]. In addition to maintaining BBB function by themselves, pericytes also promote the physiological functions of other BBB components including endothelial cells, basal lamina, and astrocytes [178][134]. For example, pericytes regulate aquaporin-4 polarization in mouse cortical astrocytes [179][135]. Furthermore, angiopoietin-1 secreted by pericytes mediates tight junction induction via the activation of Tie-2, an angiopoietin-1 receptor on EC [178,180,181][134][136][137]. Therefore, restoration of pericyte coverage may improve BBB support and promote reperfusion after stroke [182][138]. Gaining more insights into the role of pericytes in stroke could facilitate the development of novel therapeutic approaches for stroke treatment [172][128].

4.2. Pericyte Loss in Traumatic Brain Injury (TBI)

TBI, caused by an external force, is the major cause of mortality and disability, particularly in young individuals [183][139]. The secondary injury following TBI involves oxidative stress, inflammation, and the production of matrix metalloproteinases (MMPs), which contribute to BBB dysfunction [184,185,186][140][141][142]. Recent studies have highlighted pericyte degeneration as a significant factor in TBI, leading to regional microcirculatory hypoperfusion and increased BBB permeability [187,188][143][144]. A decline in pericyte markers has been observed in brain specimens from human TBI cases and in a mouse model of repetitive mild TBI up to 12 months post-injury [189][145]. Additionally, rapid pericyte loss in the acute phase of TBI has been documented in the brains of mice with TBI [188,189,190,191,192][144][145][146][147][148]. Brain pericyte apoptosis has been detected in a mouse model of TBI, suggesting that pericyte loss during TBI may be attributed to apoptosis [193][149]. It has been found that the inhibition of the TNF-α/NF-κB/iNOS axis can reverse pericyte loss, improve pericyte function, and enhance microcirculation perfusion after TBI [188][144]. This indicates the potential contribution of the TNF-α/NF-κB/iNOS axis to pericyte loss in TBI. Consequently, the development of treatments that can prevent or reverse pericyte degeneration holds promise for the management of TBI and the secondary injuries that follow.
Overall, understanding the mechanisms of pericyte loss in these conditions is crucial for developing new treatments that can prevent or reverse this process and improve patient outcomes.

References

  1. Warmke, N.; Griffin, K.J.; Cubbon, R.M. Pericytes in diabetes-associated vascular disease. J. Diabetes Complicat. 2016, 30, 1643–1650.
  2. Tu, Y.; Li, Q.; Zhou, Y.; Ye, Z.; Wu, C.; Xie, E.; Li, Y.; Li, P.; Wu, Y.; Guo, Z.; et al. Empagliflozin inhibits coronary microvascular dysfunction and reduces cardiac pericyte loss in db/db mice. Front. Cardiovasc. Med. 2022, 9, 995216.
  3. Yau, J.W.; Rogers, S.L.; Kawasaki, R.; Lamoureux, E.L.; Kowalski, J.W.; Bek, T.; Chen, S.J.; Dekker, J.M.; Fletcher, A.; Grauslund, J.; et al. Global prevalence and major risk factors of diabetic retinopathy. Diabetes Care 2012, 35, 556–564.
  4. Trost, A.; Bruckner, D.; Rivera, F.J.; Reitsamer, H.A. Pericytes in the Retina. Adv. Exp. Med. Biol. 2019, 1122, 1–26.
  5. Li, Q.; Wang, M.; Li, X.; Shao, Y. Aging and diabetic retinopathy: Inherently intertwined pathophysiological processes. Exp. Gerontol. 2023, 175, 112138.
  6. Ren, J.; Zhang, S.; Pan, Y.; Jin, M.; Li, J.; Luo, Y.; Sun, X.; Li, G. Diabetic retinopathy: Involved cells, biomarkers, and treatments. Front. Pharmacol. 2022, 13, 953691.
  7. de Oliveira, F. Pericytes in diabetic retinopathy. Br. J. Ophthalmol. 1966, 50, 134–143.
  8. Podesta, F.; Romeo, G.; Liu, W.H.; Krajewski, S.; Reed, J.C.; Gerhardinger, C.; Lorenzi, M. Bax is increased in the retina of diabetic subjects and is associated with pericyte apoptosis in vivo and in vitro. Am. J. Pathol. 2000, 156, 1025–1032.
  9. Hammes, H.P.; Lin, J.; Renner, O.; Shani, M.; Lundqvist, A.; Betsholtz, C.; Brownlee, M.; Deutsch, U. Pericytes and the pathogenesis of diabetic retinopathy. Diabetes 2002, 51, 3107–3112.
  10. Cheng, Y.; Peng, L.; Deng, X.; Li, T.; Guo, H.; Xu, C.; Fang, T.; Liu, X.; Sun, B.; Chen, L. Prostaglandin F2alpha protects against pericyte apoptosis by inhibiting the PI3K/Akt/GSK3beta/beta-catenin signaling pathway. Ann. Transl. Med. 2021, 9, 1021.
  11. Jung, E.; Kim, J.; Kim, C.S.; Kim, S.H.; Cho, M.H. Gemigliptin, a dipeptidyl peptidase-4 inhibitor, inhibits retinal pericyte injury in db/db mice and retinal neovascularization in mice with ischemic retinopathy. Biochim. Biophys. Acta 2015, 1852, 2618–2629.
  12. Engerman, R.L. Animal models of diabetic retinopathy. Trans. Sect. Ophthalmol. Am. Acad. Ophthalmol. Otolaryngol. 1976, 81, OP710–OP715.
  13. Hammes, H.P. Pericytes and the pathogenesis of diabetic retinopathy. Horm. Metab. Res. 2005, 37 (Suppl. 1), 39–43.
  14. Hammes, H.P.; Wellensiek, B.; Kloting, I.; Sickel, E.; Bretzel, R.G.; Brownlee, M. The relationship of glycaemic level to advanced glycation end-product (AGE) accumulation and retinal pathology in the spontaneous diabetic hamster. Diabetologia 1998, 41, 165–170.
  15. Hammes, H.P.; Martin, S.; Federlin, K.; Geisen, K.; Brownlee, M. Aminoguanidine treatment inhibits the development of experimental diabetic retinopathy. Proc. Natl. Acad. Sci. USA 1991, 88, 11555–11558.
  16. Tang, L.; Zhang, C.; Lu, L.; Tian, H.; Liu, K.; Luo, D.; Qiu, Q.; Xu, G.T.; Zhang, J. Melatonin Maintains Inner Blood-Retinal Barrier by Regulating Microglia via Inhibition of PI3K/Akt/Stat3/NF-kappaB Signaling Pathways in Experimental Diabetic Retinopathy. Front. Immunol. 2022, 13, 831660.
  17. Li, W.; Yanoff, M.; Liu, X.; Ye, X. Retinal capillary pericyte apoptosis in early human diabetic retinopathy. Chin. Med. J. 1997, 110, 659–663.
  18. Romeo, G.; Liu, W.H.; Asnaghi, V.; Kern, T.S.; Lorenzi, M. Activation of nuclear factor-kappaB induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes. Diabetes 2002, 51, 2241–2248.
  19. Mizutani, M.; Kern, T.S.; Lorenzi, M. Accelerated death of retinal microvascular cells in human and experimental diabetic retinopathy. J. Clin. Investig. 1996, 97, 2883–2890.
  20. Kim, Y.H.; Park, S.Y.; Park, J.; Kim, Y.S.; Hwang, E.M.; Park, J.Y.; Roh, G.S.; Kim, H.J.; Kang, S.S.; Cho, G.J.; et al. Reduction of experimental diabetic vascular leakage and pericyte apoptosis in mice by delivery of alphaA-crystallin with a recombinant adenovirus. Diabetologia 2012, 55, 2835–2844.
  21. Shin, E.S.; Huang, Q.; Gurel, Z.; Palenski, T.L.; Zaitoun, I.; Sorenson, C.M.; Sheibani, N. STAT1-mediated Bim expression promotes the apoptosis of retinal pericytes under high glucose conditions. Cell Death Dis. 2014, 5, e986.
  22. Nie, F.; Yan, J.; Ling, Y.; Liu, Z.; Fu, C.; Li, X.; Qin, Y. Effect of Shuangdan Mingmu capsule, a Chinese herbal formula, on oxidative stress-induced apoptosis of pericytes through PARP/GAPDH pathway. BMC Complement. Med. Ther. 2021, 21, 118.
  23. Chen, B.H.; Jiang, D.Y.; Tang, L.S. Advanced glycation end-products induce apoptosis involving the signaling pathways of oxidative stress in bovine retinal pericytes. Life Sci. 2006, 79, 1040–1048.
  24. Sheikpranbabu, S.; Haribalaganesh, R.; Gurunathan, S. Pigment epithelium-derived factor inhibits advanced glycation end-products-induced cytotoxicity in retinal pericytes. Diabetes Metab. 2011, 37, 505–511.
  25. Yun, J.H.; Lee, D.H.; Jeong, H.S.; Kim, S.H.; Ye, S.K.; Cho, C.H. STAT3 activation in microglia increases pericyte apoptosis in diabetic retinas through TNF-a/AKT/p70S6 kinase signaling. Biochem. Biophys. Res. Commun. 2022, 613, 133–139.
  26. Yun, J.H. Interleukin-1beta induces pericyte apoptosis via the NF-kappaB pathway in diabetic retinopathy. Biochem. Biophys. Res. Commun. 2021, 546, 46–53.
  27. Pfister, F.; Feng, Y.; vom Hagen, F.; Hoffmann, S.; Molema, G.; Hillebrands, J.L.; Shani, M.; Deutsch, U.; Hammes, H.P. Pericyte migration: A novel mechanism of pericyte loss in experimental diabetic retinopathy. Diabetes 2008, 57, 2495–2502.
  28. Lin, W.J.; Ma, X.F.; Zhou, H.R.; Xu, C.Y.; Yu, X.Y.; Hu, Y.X.; Hao, M.; Xu, Q.; Li, H.X.; Kuang, H.Y. Autophagy Modulates the Migration of Retinal Pericytes Induced by Advanced Glycation End Products. Evid. Based Complement. Altern. Med. 2022, 2022, 2760537.
  29. Monickaraj, F.; McGuire, P.; Das, A. Cathepsin D plays a role in endothelial-pericyte interactions during alteration of the blood-retinal barrier in diabetic retinopathy. FASEB J. 2018, 32, 2539–2548.
  30. McGuire, P.G.; Rangasamy, S.; Maestas, J.; Das, A. Pericyte-derived sphingosine 1-phosphate induces the expression of adhesion proteins and modulates the retinal endothelial cell barrier. Arterioscler. Thromb. Vasc. Biol. 2011, 31, e107–e115.
  31. Mendel, T.A.; Clabough, E.B.; Kao, D.S.; Demidova-Rice, T.N.; Durham, J.T.; Zotter, B.C.; Seaman, S.A.; Cronk, S.M.; Rakoczy, E.P.; Katz, A.J.; et al. Pericytes derived from adipose-derived stem cells protect against retinal vasculopathy. PLoS ONE 2013, 8, e65691.
  32. Ferland-McCollough, D.; Slater, S.; Richard, J.; Reni, C.; Mangialardi, G. Pericytes, an overlooked player in vascular pathobiology. Pharmacol. Ther. 2017, 171, 30–42.
  33. Lenoir, O.; Jasiek, M.; Henique, C.; Guyonnet, L.; Hartleben, B.; Bork, T.; Chipont, A.; Flosseau, K.; Bensaada, I.; Schmitt, A.; et al. Endothelial cell and podocyte autophagy synergistically protect from diabetes-induced glomerulosclerosis. Autophagy 2015, 11, 1130–1145.
  34. Horton, W.B.; Barrett, E.J. Microvascular Dysfunction in Diabetes Mellitus and Cardiometabolic Disease. Endocr. Rev. 2021, 42, 29–55.
  35. Khan, S.S.; Quaggin, S.E. Therapies on the Horizon for Diabetic Kidney Disease. Curr. Diab Rep. 2015, 15, 111.
  36. Kida, Y. Peritubular Capillary Rarefaction: An Underappreciated Regulator of CKD Progression. Int. J. Mol. Sci. 2020, 21, 8255.
  37. Chade, A.R. Small Vessels, Big Role: Renal Microcirculation and Progression of Renal Injury. Hypertension 2017, 69, 551–563.
  38. Lemos, D.R.; Marsh, G.; Huang, A.; Campanholle, G.; Aburatani, T.; Dang, L.; Gomez, I.; Fisher, K.; Ligresti, G.; Peti-Peterdi, J.; et al. Maintenance of vascular integrity by pericytes is essential for normal kidney function. Am. J. Physiol. Renal Physiol. 2016, 311, F1230–F1242.
  39. Kobayashi, A.; Mugford, J.W.; Krautzberger, A.M.; Naiman, N.; Liao, J.; McMahon, A.P. Identification of a multipotent self-renewing stromal progenitor population during mammalian kidney organogenesis. Stem Cell Rep. 2014, 3, 650–662.
  40. Lin, S.L.; Kisseleva, T.; Brenner, D.A.; Duffield, J.S. Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am. J. Pathol. 2008, 173, 1617–1627.
  41. Wu, C.F.; Chiang, W.C.; Lai, C.F.; Chang, F.C.; Chen, Y.T.; Chou, Y.H.; Wu, T.H.; Linn, G.R.; Ling, H.; Wu, K.D.; et al. Transforming growth factor beta-1 stimulates profibrotic epithelial signaling to activate pericyte-myofibroblast transition in obstructive kidney fibrosis. Am. J. Pathol. 2013, 182, 118–131.
  42. Schlondorff, D. The glomerular mesangial cell: An expanding role for a specialized pericyte. FASEB J. 1987, 1, 272–281.
  43. Liu, L.; Hu, X.; Cai, G.Y.; Lv, Y.; Zhuo, L.; Gao, J.J.; Cui, S.Y.; Feng, Z.; Fu, B.; Chen, X.M. High glucose-induced hypertrophy of mesangial cells is reversed by connexin43 overexpression via PTEN/Akt/mTOR signaling. Nephrol. Dial. Transplant. 2012, 27, 90–100.
  44. Pesce, C.; Menini, S.; Pricci, F.; Favre, A.; Leto, G.; DiMario, U.; Pugliese, G. Glomerular cell replication and cell loss through apoptosis in experimental diabetes mellitus. Nephron 2002, 90, 484–488.
  45. Mishra, R.; Emancipator, S.N.; Kern, T.; Simonson, M.S. High glucose evokes an intrinsic proapoptotic signaling pathway in mesangial cells. Kidney Int. 2005, 67, 82–93.
  46. Tsai, Y.C.; Kuo, M.C.; Hung, W.W.; Wu, L.Y.; Wu, P.H.; Chang, W.A.; Kuo, P.L.; Hsu, Y.L. High Glucose Induces Mesangial Cell Apoptosis through miR-15b-5p and Promotes Diabetic Nephropathy by Extracellular Vesicle Delivery. Mol. Ther. 2020, 28, 963–974.
  47. Tsai, Y.C.; Kuo, P.L.; Hung, W.W.; Wu, L.Y.; Wu, P.H.; Chang, W.A.; Kuo, M.C.; Hsu, Y.L. Angpt2 Induces Mesangial Cell Apoptosis through the MicroRNA-33-5p-SOCS5 Loop in Diabetic Nephropathy. Mol. Ther. Nucleic Acids 2018, 13, 543–555.
  48. Sharma, B.; Singh, N. Pitavastatin and 4′-hydroxy-3′-methoxyacetophenone (HMAP) reduce cognitive dysfunction in vascular dementia during experimental diabetes. Curr. Neurovascular Res. 2010, 7, 180–191.
  49. Janelidze, S.; Hertze, J.; Nagga, K.; Nilsson, K.; Nilsson, C.; Swedish Bio, F.S.G.; Wennstrom, M.; van Westen, D.; Blennow, K.; Zetterberg, H.; et al. Increased blood-brain barrier permeability is associated with dementia and diabetes but not amyloid pathology or APOE genotype. Neurobiol. Aging 2017, 51, 104–112.
  50. Stranahan, A.M.; Hao, S.; Dey, A.; Yu, X.; Baban, B. Blood-brain barrier breakdown promotes macrophage infiltration and cognitive impairment in leptin receptor-deficient mice. J. Cereb. Blood Flow. Metab. 2016, 36, 2108–2121.
  51. Lacy, M.E.; Gilsanz, P.; Eng, C.; Beeri, M.S.; Karter, A.J.; Whitmer, R.A. Severe Hypoglycemia and Cognitive Function in Older Adults with Type 1 Diabetes: The Study of Longevity in Diabetes (SOLID). Diabetes Care 2020, 43, 541–548.
  52. Kodl, C.T.; Seaquist, E.R. Cognitive dysfunction and diabetes mellitus. Endocr. Rev. 2008, 29, 494–511.
  53. Luchsinger, J.A.; Tang, M.X.; Stern, Y.; Shea, S.; Mayeux, R. Diabetes mellitus and risk of Alzheimer’s disease and dementia with stroke in a multiethnic cohort. Am. J. Epidemiol. 2001, 154, 635–641.
  54. Rom, S.; Zuluaga-Ramirez, V.; Gajghate, S.; Seliga, A.; Winfield, M.; Heldt, N.A.; Kolpakov, M.A.; Bashkirova, Y.V.; Sabri, A.K.; Persidsky, Y. Hyperglycemia-Driven Neuroinflammation Compromises BBB Leading to Memory Loss in Both Diabetes Mellitus (DM) Type 1 and Type 2 Mouse Models. Mol. Neurobiol. 2019, 56, 1883–1896.
  55. Giannini, C.; Dyck, P.J. Basement membrane reduplication and pericyte degeneration precede development of diabetic polyneuropathy and are associated with its severity. Ann. Neurol. 1995, 37, 498–504.
  56. Lin, L.; Wu, Y.; Chen, Z.; Huang, L.; Wang, L.; Liu, L. Severe Hypoglycemia Contributing to Cognitive Dysfunction in Diabetic Mice Is Associated with Pericyte and Blood-Brain Barrier Dysfunction. Front. Aging Neurosci. 2021, 13, 775244.
  57. Price, T.O.; Eranki, V.; Banks, W.A.; Ercal, N.; Shah, G.N. Topiramate treatment protects blood-brain barrier pericytes from hyperglycemia-induced oxidative damage in diabetic mice. Endocrinology 2012, 153, 362–372.
  58. Liu, Y.; Zhang, H.; Wang, S.; Guo, Y.; Fang, X.; Zheng, B.; Gao, W.; Yu, H.; Chen, Z.; Roman, R.J.; et al. Reduced pericyte and tight junction coverage in old diabetic rats are associated with hyperglycemia-induced cerebrovascular pericyte dysfunction. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H549–H562.
  59. Shah, G.N.; Price, T.O.; Banks, W.A.; Morofuji, Y.; Kovac, A.; Ercal, N.; Sorenson, C.M.; Shin, E.S.; Sheibani, N. Pharmacological inhibition of mitochondrial carbonic anhydrases protects mouse cerebral pericytes from high glucose-induced oxidative stress and apoptosis. J. Pharmacol. Exp. Ther. 2013, 344, 637–645.
  60. Price, T.O.; Sheibani, N.; Shah, G.N. Regulation of high glucose-induced apoptosis of brain pericytes by mitochondrial CA VA: A specific target for prevention of diabetic cerebrovascular pathology. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 929–935.
  61. Patrick, P.; Price, T.O.; Diogo, A.L.; Sheibani, N.; Banks, W.A.; Shah, G.N. Topiramate Protects Pericytes from Glucotoxicity: Role for Mitochondrial CA VA in Cerebromicrovascular Disease in Diabetes. J. Endocrinol. Diabetes 2015, 2.
  62. May, J.M.; Jayagopal, A.; Qu, Z.C.; Parker, W.H. Ascorbic acid prevents high glucose-induced apoptosis in human brain pericytes. Biochem. Biophys. Res. Commun. 2014, 452, 112–117.
  63. Kannel, W.B.; Hjortland, M.; Castelli, W.P. Role of diabetes in congestive heart failure: The Framingham study. Am. J. Cardiol. 1974, 34, 29–34.
  64. Stamler, J.; Vaccaro, O.; Neaton, J.D.; Wentworth, D. Diabetes, other risk factors, and 12-yr cardiovascular mortality for men screened in the Multiple Risk Factor Intervention Trial. Diabetes Care 1993, 16, 434–444.
  65. Yoon, Y.S.; Uchida, S.; Masuo, O.; Cejna, M.; Park, J.S.; Gwon, H.C.; Kirchmair, R.; Bahlman, F.; Walter, D.; Curry, C.; et al. Progressive attenuation of myocardial vascular endothelial growth factor expression is a seminal event in diabetic cardiomyopathy: Restoration of microvascular homeostasis and recovery of cardiac function in diabetic cardiomyopathy after replenishment of local vascular endothelial growth factor. Circulation 2005, 111, 2073–2085.
  66. Wakisaka, M.; Kamouchi, M.; Kitazono, T. Lessons from the Trials for the Desirable Effects of Sodium Glucose Co-Transporter 2 Inhibitors on Diabetic Cardiovascular Events and Renal Dysfunction. Int. J. Mol. Sci. 2019, 20, 5668.
  67. Regan, T.J.; Lyons, M.M.; Ahmed, S.S.; Levinson, G.E.; Oldewurtel, H.A.; Ahmad, M.R.; Haider, B. Evidence for cardiomyopathy in familial diabetes mellitus. J. Clin. Investig. 1977, 60, 884–899.
  68. Kawaguchi, M.; Techigawara, M.; Ishihata, T.; Asakura, T.; Saito, F.; Maehara, K.; Maruyama, Y. A comparison of ultrastructural changes on endomyocardial biopsy specimens obtained from patients with diabetes mellitus with and without hypertension. Heart Vessel. 1997, 12, 267–274.
  69. Hinkel, R.; Howe, A.; Renner, S.; Ng, J.; Lee, S.; Klett, K.; Kaczmarek, V.; Moretti, A.; Laugwitz, K.L.; Skroblin, P.; et al. Diabetes Mellitus-Induced Microvascular Destabilization in the Myocardium. J. Am. Coll. Cardiol. 2017, 69, 131–143.
  70. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562.
  71. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150.
  72. Iadecola, C.; Duering, M.; Hachinski, V.; Joutel, A.; Pendlebury, S.T.; Schneider, J.A.; Dichgans, M. Vascular Cognitive Impairment and Dementia: JACC Scientific Expert Panel. J. Am. Coll. Cardiol. 2019, 73, 3326–3344.
  73. Sagare, A.P.; Bell, R.D.; Zhao, Z.; Ma, Q.; Winkler, E.A.; Ramanathan, A.; Zlokovic, B.V. Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nat. Commun. 2013, 4, 2932.
  74. Ding, R.; Hase, Y.; Ameen-Ali, K.E.; Ndung’u, M.; Stevenson, W.; Barsby, J.; Gourlay, R.; Akinyemi, T.; Akinyemi, R.; Uemura, M.T.; et al. Loss of capillary pericytes and the blood-brain barrier in white matter in poststroke and vascular dementias and Alzheimer’s disease. Brain Pathol. 2020, 30, 1087–1101.
  75. Montagne, A.; Nikolakopoulou, A.M.; Zhao, Z.; Sagare, A.P.; Si, G.; Lazic, D.; Barnes, S.R.; Daianu, M.; Ramanathan, A.; Go, A.; et al. Pericyte degeneration causes white matter dysfunction in the mouse central nervous system. Nat. Med. 2018, 24, 326–337.
  76. Miners, J.S.; Schulz, I.; Love, S. Differing associations between Abeta accumulation, hypoperfusion, blood-brain barrier dysfunction and loss of PDGFRB pericyte marker in the precuneus and parietal white matter in Alzheimer’s disease. J. Cereb. Blood Flow. Metab. 2018, 38, 103–115.
  77. Sengillo, J.D.; Winkler, E.A.; Walker, C.T.; Sullivan, J.S.; Johnson, M.; Zlokovic, B.V. Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer’s disease. Brain Pathol. 2013, 23, 303–310.
  78. Li, P.; Wu, Y.; Hamlett, E.D.; Goodwin, A.J.; Halushka, P.V.; Carroll, S.L.; Liu, M.; Fan, H. Suppression of Fli-1 protects against pericyte loss and cognitive deficits in Alzheimer’s disease. Mol. Ther. 2022, 30, 1451–1464.
  79. Shi, H.; Koronyo, Y.; Rentsendorj, A.; Regis, G.C.; Sheyn, J.; Fuchs, D.T.; Kramerov, A.A.; Ljubimov, A.V.; Dumitrascu, O.M.; Rodriguez, A.R.; et al. Identification of early pericyte loss and vascular amyloidosis in Alzheimer’s disease retina. Acta Neuropathol. 2020, 139, 813–836.
  80. Janota, C.S.; Brites, D.; Lemere, C.A.; Brito, M.A. Glio-vascular changes during ageing in wild-type and Alzheimer’s disease-like APP/PS1 mice. Brain Res. 2015, 1620, 153–168.
  81. Wu, Q.; Yuan, X.; Bai, J.; Han, R.; Li, Z.; Zhang, H.; Xiu, R. MicroRNA-181a protects against pericyte apoptosis via directly targeting FOXO1: Implication for ameliorated cognitive deficits in APP/PS1 mice. Aging 2019, 11, 6120–6133.
  82. Shi, H.; Koronyo, Y.; Fuchs, D.T.; Sheyn, J.; Wawrowsky, K.; Lahiri, S.; Black, K.L.; Koronyo-Hamaoui, M. Retinal capillary degeneration and blood-retinal barrier disruption in murine models of Alzheimer’s disease. Acta Neuropathol. Commun. 2020, 8, 202.
  83. Kiernan, M.C.; Vucic, S.; Cheah, B.C.; Turner, M.R.; Eisen, A.; Hardiman, O.; Burrell, J.R.; Zoing, M.C. Amyotrophic lateral sclerosis. Lancet 2011, 377, 942–955.
  84. Al-Chalabi, A.; Hardiman, O. The epidemiology of ALS: A conspiracy of genes, environment and time. Nat. Rev. Neurol. 2013, 9, 617–628.
  85. Sasaki, S. Alterations of the blood-spinal cord barrier in sporadic amyotrophic lateral sclerosis. Neuropathology 2015, 35, 518–528.
  86. Cao, M.C.; Cawston, E.E.; Chen, G.; Brooks, C.; Douwes, J.; McLean, D.; Graham, E.S.; Dragunow, M.; Scotter, E.L. Serum biomarkers of neuroinflammation and blood-brain barrier leakage in amyotrophic lateral sclerosis. BMC Neurol. 2022, 22, 216.
  87. Coatti, G.C.; Cavacana, N.; Zatz, M. The Role of Pericytes in Amyotrophic Lateral Sclerosis. Adv. Exp. Med. Biol. 2019, 1147, 137–146.
  88. Yamadera, M.; Fujimura, H.; Inoue, K.; Toyooka, K.; Mori, C.; Hirano, H.; Sakoda, S. Microvascular disturbance with decreased pericyte coverage is prominent in the ventral horn of patients with amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2015, 16, 393–401.
  89. Winkler, E.A.; Sengillo, J.D.; Sullivan, J.S.; Henkel, J.S.; Appel, S.H.; Zlokovic, B.V. Blood-spinal cord barrier breakdown and pericyte reductions in amyotrophic lateral sclerosis. Acta Neuropathol. 2013, 125, 111–120.
  90. Saul, J.; Hutchins, E.; Reiman, R.; Saul, M.; Ostrow, L.W.; Harris, B.T.; Van Keuren-Jensen, K.; Bowser, R.; Bakkar, N. Global alterations to the choroid plexus blood-CSF barrier in amyotrophic lateral sclerosis. Acta Neuropathol. Commun. 2020, 8, 92.
  91. Garbuzova-Davis, S.; Boccio, K.J.; Llauget, A.; Shell, R.; Hailu, S.; Mustafa, H.; Ehrhart, J.; Sanberg, P.R.; Appel, S.H.; Borlongan, C.V. Beneficial Effects of Transplanted Human Bone Marrow Endothelial Progenitors on Functional and Cellular Components of Blood-Spinal Cord Barrier in ALS Mice. Eneuro 2021, 8, ENEURO.0314-21.2021.
  92. Coatti, G.C.; Frangini, M.; Valadares, M.C.; Gomes, J.P.; Lima, N.O.; Cavacana, N.; Assoni, A.F.; Pelatti, M.V.; Birbrair, A.; de Lima, A.C.P.; et al. Pericytes Extend Survival of ALS SOD1 Mice and Induce the Expression of Antioxidant Enzymes in the Murine Model and in IPSCs Derived Neuronal Cells from an ALS Patient. Stem Cell Rev. Rep. 2017, 13, 686–698.
  93. Dolin, H.H.; Papadimos, T.J.; Stepkowski, S.; Chen, X.; Pan, Z.K. A Novel Combination of Biomarkers to Herald the Onset of Sepsis Prior to the Manifestation of Symptoms. Shock 2018, 49, 364–370.
  94. Hotchkiss, R.S.; Moldawer, L.L.; Opal, S.M.; Reinhart, K.; Turnbull, I.R.; Vincent, J.L. Sepsis and septic shock. Nat. Rev. Dis. Primers 2016, 2, 16045.
  95. Li, P.; Zhou, Y.; Goodwin, A.J.; Cook, J.A.; Halushka, P.V.; Zhang, X.K.; Wilson, C.L.; Schnapp, L.M.; Zingarelli, B.; Fan, H. Fli-1 Governs Pericyte Dysfunction in a Murine Model of Sepsis. J. Infect. Dis. 2018, 218, 1995–2005.
  96. Stasi, A.; Franzin, R.; Divella, C.; Gesualdo, L.; Stallone, G.; Castellano, G. Double Labeling of PDGFR-beta and alpha-SMA in Swine Models of Acute Kidney Injury to Detect Pericyte-to-Myofibroblast Transdifferentation as Early Marker of Fibrosis. Bio Protoc. 2020, 10, e3779.
  97. Nishioku, T.; Dohgu, S.; Takata, F.; Eto, T.; Ishikawa, N.; Kodama, K.B.; Nakagawa, S.; Yamauchi, A.; Kataoka, Y. Detachment of brain pericytes from the basal lamina is involved in disruption of the blood-brain barrier caused by lipopolysaccharide-induced sepsis in mice. Cell Mol. Neurobiol. 2009, 29, 309–316.
  98. Barichello, T.; Generoso, J.S.; Collodel, A.; Petronilho, F.; Dal-Pizzol, F. The blood-brain barrier dysfunction in sepsis. Tissue Barriers 2021, 9, 1840912.
  99. Armulik, A.; Genove, G.; Mae, M.; Nisancioglu, M.H.; Wallgard, E.; Niaudet, C.; He, L.; Norlin, J.; Lindblom, P.; Strittmatter, K.; et al. Pericytes regulate the blood-brain barrier. Nature 2010, 468, 557–561.
  100. Zeng, H.; He, X.; Tuo, Q.H.; Liao, D.F.; Zhang, G.Q.; Chen, J.X. LPS causes pericyte loss and microvascular dysfunction via disruption of Sirt3/angiopoietins/Tie-2 and HIF-2alpha/Notch3 pathways. Sci. Rep. 2016, 6, 20931.
  101. Zhang, Z.S.; Liu, Y.Y.; He, S.S.; Bao, D.Q.; Wang, H.C.; Zhang, J.; Peng, X.Y.; Zang, J.T.; Zhu, Y.; Wu, Y.; et al. Pericytes protect rats and mice from sepsis-induced injuries by maintaining vascular reactivity and barrier function: Implication of miRNAs and microvesicles. Mil. Med. Res. 2023, 10, 13.
  102. Lindl, K.A.; Marks, D.R.; Kolson, D.L.; Jordan-Sciutto, K.L. HIV-associated neurocognitive disorder: Pathogenesis and therapeutic opportunities. J. Neuroimmune Pharmacol. 2010, 5, 294–309.
  103. Castro, V.; Bertrand, L.; Luethen, M.; Dabrowski, S.; Lombardi, J.; Morgan, L.; Sharova, N.; Stevenson, M.; Blasig, I.E.; Toborek, M. Occludin controls HIV transcription in brain pericytes via regulation of SIRT-1 activation. FASEB J. 2016, 30, 1234–1246.
  104. Nakagawa, S.; Castro, V.; Toborek, M. Infection of human pericytes by HIV-1 disrupts the integrity of the blood-brain barrier. J. Cell Mol. Med. 2012, 16, 2950–2957.
  105. Torices, S.; Roberts, S.A.; Park, M.; Malhotra, A.; Toborek, M. Occludin, caveolin-1, and Alix form a multi-protein complex and regulate HIV-1 infection of brain pericytes. FASEB J. 2020, 34, 16319–16332.
  106. Persidsky, Y.; Hill, J.; Zhang, M.; Dykstra, H.; Winfield, M.; Reichenbach, N.L.; Potula, R.; Mukherjee, A.; Ramirez, S.H.; Rom, S. Dysfunction of brain pericytes in chronic neuroinflammation. J. Cereb. Blood Flow. Metab. 2016, 36, 794–807.
  107. Cho, H.J.; Kuo, A.M.; Bertrand, L.; Toborek, M. HIV Alters Gap Junction-Mediated Intercellular Communication in Human Brain Pericytes. Front. Mol. Neurosci. 2017, 10, 410.
  108. Naranjo, O.; Torices, S.; Clifford, P.R.; Daftari, M.T.; Osborne, O.M.; Fattakhov, N.; Toborek, M. Pericyte infection by HIV-1: A fatal attraction. Retrovirology 2022, 19, 27.
  109. Niu, F.; Yao, H.; Zhang, W.; Sutliff, R.L.; Buch, S. Tat 101-mediated enhancement of brain pericyte migration involves platelet-derived growth factor subunit B homodimer: Implications for human immunodeficiency virus-associated neurocognitive disorders. J. Neurosci. 2014, 34, 11812–11825.
  110. Bohannon, D.G.; Ko, A.; Filipowicz, A.R.; Kuroda, M.J.; Kim, W.K. Dysregulation of sonic hedgehog pathway and pericytes in the brain after lentiviral infection. J. Neuroinflamm. 2019, 16, 86.
  111. Niu, F.; Yao, H.; Liao, K.; Buch, S. HIV Tat 101-mediated loss of pericytes at the blood-brain barrier involves PDGF-BB. Ther. Targets Neurol. Dis. 2015, 2, e471.
  112. Burkert, F.R.; Lanser, L.; Bellmann-Weiler, R.; Weiss, G. Coronavirus Disease 2019: Clinics, Treatment, and Prevention. Front. Microbiol. 2021, 12, 761887.
  113. Avolio, E.; Carrabba, M.; Milligan, R.; Kavanagh Williamson, M.; Beltrami, A.P.; Gupta, K.; Elvers, K.T.; Gamez, M.; Foster, R.R.; Gillespie, K.; et al. The SARS-CoV-2 Spike protein disrupts human cardiac pericytes function through CD147 receptor-mediated signalling: A potential non-infective mechanism of COVID-19 microvascular disease. Clin. Sci. 2021, 135, 2667–2689.
  114. Bocci, M.; Oudenaarden, C.; Saenz-Sarda, X.; Simren, J.; Eden, A.; Sjolund, J.; Moller, C.; Gisslen, M.; Zetterberg, H.; Englund, E.; et al. Infection of Brain Pericytes Underlying Neuropathology of COVID-19 Patients. Int. J. Mol. Sci. 2021, 22, 11622.
  115. Khan, A.O.; Reyat, J.S.; Hill, H.; Bourne, J.H.; Colicchia, M.; Newby, M.L.; Allen, J.D.; Crispin, M.; Youd, E.; Murray, P.G.; et al. Preferential uptake of SARS-CoV-2 by pericytes potentiates vascular damage and permeability in an organoid model of the microvasculature. Cardiovasc. Res. 2022, 118, 3085–3096.
  116. Das, M.; Chung, M.K. Cardiac Pericytes: Underappreciated Targets of SARS-CoV-2. JACC Basic Transl. Sci. 2023, 8, 121–123.
  117. Nicin, L.; Abplanalp, W.T.; Mellentin, H.; Kattih, B.; Tombor, L.; John, D.; Schmitto, J.D.; Heineke, J.; Emrich, F.; Arsalan, M.; et al. Cell type-specific expression of the putative SARS-CoV-2 receptor ACE2 in human hearts. Eur. Heart J. 2020, 41, 1804–1806.
  118. Brumback, B.D.; Dmytrenko, O.; Robinson, A.N.; Bailey, A.L.; Ma, P.; Liu, J.; Hicks, S.C.; Ng, S.; Li, G.; Zhang, D.M.; et al. Human Cardiac Pericytes Are Susceptible to SARS-CoV-2 Infection. JACC Basic Transl. Sci. 2023, 8, 109–120.
  119. Robinson, F.A.; Mihealsick, R.P.; Wagener, B.M.; Hanna, P.; Poston, M.D.; Efimov, I.R.; Shivkumar, K.; Hoover, D.B. Role of angiotensin-converting enzyme 2 and pericytes in cardiac complications of COVID-19 infection. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H1059–H1068.
  120. Daems, M.; Liesenborghs, L.; Boudewijns, R.; Simmonds, S.J.; Kraisin, S.; Van Wauwe, J.; Cuijpers, I.; Raman, J.; Geuens, N.; Buyten, T.V.; et al. SARS-CoV-2 infection causes prolonged cardiomyocyte swelling and inhibition of HIF1alpha translocation in an animal model COVID-19. Front. Cardiovasc. Med. 2022, 9, 964512.
  121. Muhl, L.; He, L.; Sun, Y.; Andaloussi Mae, M.; Pietila, R.; Liu, J.; Genove, G.; Zhang, L.; Xie, Y.; Leptidis, S.; et al. The SARS-CoV-2 receptor ACE2 is expressed in mouse pericytes but not endothelial cells: Implications for COVID-19 vascular research. Stem Cell Rep. 2022, 17, 1089–1104.
  122. Miners, J.S.; Fisher, R.A.; Love, S. SARS-CoV-2 targets pericytes to restrict blood flow within the brain. Brain 2023, 146, 418–420.
  123. Khaddaj-Mallat, R.; Aldib, N.; Bernard, M.; Paquette, A.S.; Ferreira, A.; Lecordier, S.; Saghatelyan, A.; Flamand, L.; ElAli, A. SARS-CoV-2 deregulates the vascular and immune functions of brain pericytes via Spike protein. Neurobiol. Dis. 2021, 161, 105561.
  124. Ju, J.; Su, Y.; Zhou, Y.; Wei, H.; Xu, Q. The SARS-CoV-2 envelope protein disrupts barrier function in an in vitro human blood-brain barrier model. Front. Cell Neurosci. 2022, 16, 897564.
  125. Cardot-Leccia, N.; Hubiche, T.; Dellamonica, J.; Burel-Vandenbos, F.; Passeron, T. Pericyte alteration sheds light on micro-vasculopathy in COVID-19 infection. Intensive Care Med. 2020, 46, 1777–1778.
  126. Jones, O.Y.; Yeralan, S. Is Long COVID a State of Systemic Pericyte Disarray? J. Clin. Med. 2022, 11, 572.
  127. Uemura, M.T.; Maki, T.; Ihara, M.; Lee, V.M.Y.; Trojanowski, J.Q. Brain Microvascular Pericytes in Vascular Cognitive Impairment and Dementia. Front. Aging Neurosci. 2020, 12, 80.
  128. Cao, L.; Zhou, Y.; Chen, M.; Li, L.; Zhang, W. Pericytes for Therapeutic Approaches to Ischemic Stroke. Front. Neurosci. 2021, 15, 629297.
  129. Fernandez-Klett, F.; Potas, J.R.; Hilpert, D.; Blazej, K.; Radke, J.; Huck, J.; Engel, O.; Stenzel, W.; Genove, G.; Priller, J. Early loss of pericytes and perivascular stromal cell-induced scar formation after stroke. J. Cereb. Blood Flow. Metab. 2013, 33, 428–439.
  130. Zhang, Y.; Zhang, X.; Wei, Q.; Leng, S.; Li, C.; Han, B.; Bai, Y.; Zhang, H.; Yao, H. Activation of Sigma-1 Receptor Enhanced Pericyte Survival via the Interplay between Apoptosis and Autophagy: Implications for Blood-Brain Barrier Integrity in Stroke. Transl. Stroke Res. 2020, 11, 267–287.
  131. Tsao, C.C.; Baumann, J.; Huang, S.F.; Kindler, D.; Schroeter, A.; Kachappilly, N.; Gassmann, M.; Rudin, M.; Ogunshola, O.O. Pericyte hypoxia-inducible factor-1 (HIF-1) drives blood-brain barrier disruption and impacts acute ischemic stroke outcome. Angiogenesis 2021, 24, 823–842.
  132. Zhou, Y.F.; Li, P.C.; Wu, J.H.; Haslam, J.A.; Mao, L.; Xia, Y.P.; He, Q.W.; Wang, X.X.; Lei, H.; Lan, X.L.; et al. Sema3E/PlexinD1 inhibition is a therapeutic strategy for improving cerebral perfusion and restoring functional loss after stroke in aged rats. Neurobiol. Aging 2018, 70, 102–116.
  133. Ozen, I.; Roth, M.; Barbariga, M.; Gaceb, A.; Deierborg, T.; Genove, G.; Paul, G. Loss of Regulator of G-Protein Signaling 5 Leads to Neurovascular Protection in Stroke. Stroke 2018, 49, 2182–2190.
  134. Cai, W.; Liu, H.; Zhao, J.; Chen, L.Y.; Chen, J.; Lu, Z.; Hu, X. Pericytes in Brain Injury and Repair After Ischemic Stroke. Transl. Stroke Res. 2017, 8, 107–121.
  135. Gundersen, G.A.; Vindedal, G.F.; Skare, O.; Nagelhus, E.A. Evidence that pericytes regulate aquaporin-4 polarization in mouse cortical astrocytes. Brain Struct. Funct. 2014, 219, 2181–2186.
  136. Hori, S.; Ohtsuki, S.; Hosoya, K.; Nakashima, E.; Terasaki, T. A pericyte-derived angiopoietin-1 multimeric complex induces occludin gene expression in brain capillary endothelial cells through Tie-2 activation in vitro. J. Neurochem. 2004, 89, 503–513.
  137. Sundberg, C.; Kowanetz, M.; Brown, L.F.; Detmar, M.; Dvorak, H.F. Stable expression of angiopoietin-1 and other markers by cultured pericytes: Phenotypic similarities to a subpopulation of cells in maturing vessels during later stages of angiogenesis in vivo. Lab. Investig. 2002, 82, 387–401.
  138. van Dijk, C.G.; Nieuweboer, F.E.; Pei, J.Y.; Xu, Y.J.; Burgisser, P.; van Mulligen, E.; el Azzouzi, H.; Duncker, D.J.; Verhaar, M.C.; Cheng, C. The complex mural cell: Pericyte function in health and disease. Int. J. Cardiol. 2015, 190, 75–89.
  139. Menon, D.K.; Schwab, K.; Wright, D.W.; Maas, A.I. Demographics and Clinical Assessment Working Group of the International and Interagency Initiative toward Common Data Elements for Research on Traumatic Brain Injury and Psychological Health. Position statement: Definition of traumatic brain injury. Arch. Phys. Med. Rehabil. 2010, 91, 1637–1640.
  140. Ladak, A.A.; Enam, S.A.; Ibrahim, M.T. A Review of the Molecular Mechanisms of Traumatic Brain Injury. World Neurosurg. 2019, 131, 126–132.
  141. Abdul-Muneer, P.M.; Long, M.; Conte, A.A.; Santhakumar, V.; Pfister, B.J. High Ca(2+) Influx During Traumatic Brain Injury Leads to Caspase-1-Dependent Neuroinflammation and Cell Death. Mol. Neurobiol. 2017, 54, 3964–3975.
  142. Patel, R.K.; Prasad, N.; Kuwar, R.; Haldar, D.; Abdul-Muneer, P.M. Transforming growth factor-beta 1 signaling regulates neuroinflammation and apoptosis in mild traumatic brain injury. Brain Behav. Immun. 2017, 64, 244–258.
  143. Dore-Duffy, P.; Wang, S.; Mehedi, A.; Katyshev, V.; Cleary, K.; Tapper, A.; Reynolds, C.; Ding, Y.; Zhan, P.; Rafols, J.; et al. Pericyte-mediated vasoconstriction underlies TBI-induced hypoperfusion. Neurol. Res. 2011, 33, 176–186.
  144. Zheng, S.; Wang, C.; Lin, L.; Mu, S.; Liu, H.; Hu, X.; Chen, X.; Wang, S. TNF-alpha Impairs Pericyte-Mediated Cerebral Microcirculation via the NF-kappaB/iNOS Axis after Experimental Traumatic Brain Injury. J. Neurotrauma 2023, 40, 349–364.
  145. Ojo, J.; Eisenbaum, M.; Shackleton, B.; Lynch, C.; Joshi, U.; Saltiel, N.; Pearson, A.; Ringland, C.; Paris, D.; Mouzon, B.; et al. Mural cell dysfunction leads to altered cerebrovascular tau uptake following repetitive head trauma. Neurobiol. Dis. 2021, 150, 105237.
  146. Zehendner, C.M.; Sebastiani, A.; Hugonnet, A.; Bischoff, F.; Luhmann, H.J.; Thal, S.C. Traumatic brain injury results in rapid pericyte loss followed by reactive pericytosis in the cerebral cortex. Sci. Rep. 2015, 5, 13497.
  147. Bhowmick, S.; D’Mello, V.; Caruso, D.; Wallerstein, A.; Abdul-Muneer, P.M. Impairment of pericyte-endothelium crosstalk leads to blood-brain barrier dysfunction following traumatic brain injury. Exp. Neurol. 2019, 317, 260–270.
  148. Wu, Y.; Wu, H.; Zeng, J.; Pluimer, B.; Dong, S.; Xie, X.; Guo, X.; Ge, T.; Liang, X.; Feng, S.; et al. Mild traumatic brain injury induces microvascular injury and accelerates Alzheimer-like pathogenesis in mice. Acta Neuropathol. Commun. 2021, 9, 74.
  149. Choi, Y.K.; Maki, T.; Mandeville, E.T.; Koh, S.H.; Hayakawa, K.; Arai, K.; Kim, Y.M.; Whalen, M.J.; Xing, C.; Wang, X.; et al. Dual effects of carbon monoxide on pericytes and neurogenesis in traumatic brain injury. Nat. Med. 2016, 22, 1335–1341.
More
Video Production Service