Summary

Isolation of Mouse Pancreatic Endothelial Cells

Published: June 21, 2024
doi:

Summary

This protocol describes the isolation of mouse endothelial cells from whole pancreas.

Abstract

The pancreas is a vital organ for maintaining metabolic balance within the body, in part due to its production of metabolic hormones such as insulin and glucagon, as well as digestive enzymes. The pancreas is also a highly vascularized organ, a feature facilitated by the intricate network of pancreatic capillaries. This extensive capillary network is made up of highly fenestrated endothelial cells (ECs) important for pancreas development and function. Accordingly, the dysfunction of ECs can contribute to that of the pancreas in diseases like diabetes and cancer. Thus, researching the function of pancreatic ECs (pECs) is important not only for understanding pancreas biology but also for developing its pathologies. Mouse models are valuable tools to study metabolic and cardiovascular diseases. However, there has not been an established protocol with sufficient details described for the isolation of mouse pECs due to the relatively small population of ECs and the abundant digestive enzymes potentially released from the acinar tissue that can lead to cell damage and, thus, low yield. To address these challenges, we devised a protocol to enrich and recover mouse pECs, combining gentle physical and chemical dissociation and antibody-mediated selection. The protocol presented here provides a robust method to extract intact and viable ECs from the whole mouse pancreas. This protocol is suitable for multiple downstream assays and may be applied to various mouse models.

Introduction

The pancreas, key to metabolic control and homeostasis, is a highly vascularized organ. The pancreas has both endocrine and exocrine functions, controlling the regulation of blood glucose and digestive enzymes, respectively. These two compartments are linked together by the extensive network of pancreatic blood vessels, facilitating the exchange and transport of oxygen, hormones, and enzymes. Critically, this dense capillary network penetrates the Islet of Langerhans, a cluster of hormone-regulating cells within the pancreas responsible for its endocrine function, consisting of the glucagon-secreting alpha (α) cells, the insulin-secreting beta (β) cells and the somatostatin-secreting delta (δ) cells1,2. Although the islets only make up 1-2% of the pancreatic mass, they receive 20% of total blood flow3, highlighting the importance of islet vasculature. The pancreatic capillaries are primarily made up of highly fenestrated endothelial cells (ECs), which are surrounded by mural pericytes. These capillary ECs play a vital role in the islet development, maturation, and (dys)function and form intimate crosstalks with various endo- and exocrine cells4 (Figure 1).

Endothelial dysfunction has been observed in both Type 1 and Type 2 diabetes, the most common conditions caused by pancreatic islet dysfunction5,6. Both islet microvascular density and morphology can be altered in diabetes7. Moreover, pancreatic cancer, a highly aggressive tumor that can also be manifested as diabetes, is characterized by high microvascular density with poor perfusion8. Given the pivotal structural and functional roles of ECs in both normal and diseased pancreatic tissue, there is a pertinent need to study the pECs in development, physiology, and pathology to unveil the mechanisms that drive health or diseases.

Numerous protocols have been developed for the isolation of ECs from different murine (e.g., brain9,10, lung11, heart12, liver13, skeletal muscles14, and adipose tissues15) and human (e.g., brain16, visceral adipose tissue17,18, peripheral nerves19, lung20,21,22, and mesenteric artery23) tissues. These protocols typically involve the use of enzymatic digestions (e.g., by collagenase, trypsin24, dispase24,25, and liberase26), followed by an antibody-based enrichment step. Moreover, these protocols tend to rely on extended durations of digestions in high concentrations of enzymes with vigorous agitation at 37 °C (Table 1). Due to the unique features of the pancreas, including that it houses a plethora of endogenous digestive enzymes, these existing protocols cannot be directly applied to isolate pECs. First, the extracellular matrix (ECM) composition of the pancreas is different from other tissues. While collagenase is commonly used for EC isolation, there are multiple subtypes with different tissue-specific dissociation capabilities, thus requiring optimization. Second, and crucial to pEC isolation, the release and activation of pancreatic endogenous enzymes can significantly hinder the isolation process. To this end, caution needs to be taken to minimize the rupture of the exocrine acinar cells (the primary source of zymogens, proteases, and RNase27), which can induce further cell damage and result in low cell viability and overall affect recovery27,28,29.

To address these challenges, we have adapted methods from existing EC isolation protocols and established a new protocol suitable for EC isolation from mouse pancreases. Specifically, we describe here a workflow (Figure 2) using collagenase Type I (typically implemented for lung EC isolation), lower digestion temperatures and no agitation (to prevent activation of pancreatic zymogens), and DNase30,31,32 supplementation (to prevent DNA-induced apoptosis and improve cell viability, and an antibody for CD3133 (PECAM1, a pan-EC marker). The described protocol produces EC populations isolated from mouse pancreas that can be used for gene expression profiling and protein assays.

Protocol

Tissue isolation was performed under the approved study protocol #17010 by the Institutional Animal Care and Use Committee (IACUC) of Beckman Research Institute, City of Hope (Duarte, California, USA). Here, we use Tie-2CreERT2;Rosa26-TdTomato mouse line in C57BL/6 background at 8 – 12 weeks of age. In this line, ECs are labeled with TdTomato when induced with tamoxifen as previously described34. However, this protocol can be adapted for all ages of adult mice with different genotypes and genetic …

Representative Results

Following this protocol, approximately 2 x 106 live cells can be obtained when pooling 3 mouse pancreases, and 750,000 cells from a single mouse pancreas. To validate the enrichment of EC, we performed the following analyses: 1) quantitative PCR: compared to the flow-through (FT) samples (i.e., the non-CD31 antibody-bound fractions), the EC fractions had significantly higher levels of Pecam1 (encoding CD31) and Kdr (encoding VEGFR2), two EC marker genes33, and lower le…

Discussion

In this article, we present a protocol for enrichment and isolation of the pECs. Similar to previous EC isolation protocols from other tissues or organs, this protocol consists of three major processes, namely, physical dissociation, enzymatic digestion, and antibody-based EC enrichment. To address the unique challenges in processing the pancreas, we introduced several key adaptations and critical steps within our protocol: 1) a gentle one-step collagenase digestion with a short incubation time, 2) supplementation of hig…

Offenlegungen

The authors have nothing to disclose.

Acknowledgements

The authors thank Dr. Brian Armstrong at City of Hope, and Mindy Rodriguez at University of California, Riverside for technical assistance. This study was funded in part by grants from the NIH (R01 HL145170 to ZBC), Ella Fitzgerald Foundation (to ZBC), City of Hope (Arthur Riggs Diabetes Metabolism and Research Institute Innovation Award), and California Institute of Regenerative Medicine grant EDU4-12772 (to AT). Research reported in this publication included work performed in the Light Microscopy and Digital Imaging supported by the National Cancer Institute of the NIH under award number P30CA033572. Figure 1 and Figure 2 were made with BioRender.

Materials

1.5 mL eppendorf USA Scientific 1615-5500
10 cm dish Genesee Scientific 25-202
25G needles BD 305145
2X Taq Pro Universal SYBR qPCR Master Mix Vazyme Q712-03-AA
5 mL eppendorf Thermo Fisher  14282300
6-well plate Greiner Bio-One 07-000-208
70 µm strainer Fisher 22-363-548
Anti-CD31-biotin Miltenyi Biotech REA784
Bovine serum albumin heat shock treated Fisher BP1600-100
CaCl2 Fisher BP510
Centrifuge Eppendorf
Collagen Type 1, from calf skin Sigma Aldrich  C9791 Attachment reagent in the protocol
Collagenase Type 1  Worthington Bio LS004197
Countess Automatic Cell Counter Thermo Fisher 
DAPI Thermo Fisher  D1306 immunofluorescence
Disposable Safety Scalpels Myco Instrumentation 6008TR-10
DNAse I  Roche 260913 
D-PBS (Ca2+,Mg2+) Thermo Fisher  14080055
Ethanol Fisher BP2818-4
Fetal bovine serum Fisher 10437028
Incubator Kept at 37 °C 5% CO2
LS Columns Miltenyi Biotech 130-042-401
M199 Sigma M2520-1L
MACS MultiStand with the QuadroMACS Separator  Miltenyi Biotech 130-042-303
Medium 199 Sigma Aldrich  M2520-10X
Microbeads anti-biotin Miltenyi Biotech 130-090-485
Microscope Leica To assess cell morphology
Molecular Grade Water Corning 46-000-CM
NaCl Fisher S271-1
New Brunswick Innova 44/44R Orbital shaker  Eppendorf
PECAM1 (CD31) Antibody Abcam ab56299 immunofluorescence
PECAM1 (CD31) Antibody R&D Systems AF3628
Phosphate Buffered Saline (10X) (no Ca2+,no Mg2+) Genesee Scientific 25-507-XB
Primer 36B4 Forward mouse IDT AGATTCGGGATATGCTGTTGGC
Primer 36B4 Revese mouse  IDT TCGGGTCCTAGACCAGTGTTC
Primer Kdr Forward mouse  IDT TCCAGAATCCTCTTCCATGC
Primer Kdr Reverse mouse IDT AAACCTCCTGCAAGCAAATG
Primer Nkx6.1 Reverse mouse  IDT CACGGCGGACTCTGCATCACTC
Primer Nxk6.1 Forward mouse IDT CTCTACTTTAGCCCCAGCG
Primer PECAM1 Forward mouse IDT ACGCTGGTGCTCTATGCAAG
Primer PECAM1 Reverse mouse IDT TCAGTTGCTGCCCATTCATCA
RNase ZAP Thermo Fisher  AM9780
RNase-free water Takara RR036B
Sterile 12" long forceps F.S.T 91100-16
Sterile fine forceps F.S.T 11050-10
Sterile fine scissors F.S.T 14061-11
Tissue Culture Dishes 2cm Genesee Scientific 25-260
TRIzol reagent Fisher 15596018
Trypan Blue Corning MT25900CI
Trypsin Inhibitor  Roche 10109886001
Tween-20
VE-Cadherin Antibody Abcam ab33168 immunofluorescence
Waterbath

Referenzen

  1. Ballian, N., Brunicardi, F. C. Islet vasculature as a regulator of endocrine pancreas function. World J Surg. 31 (4), 705-714 (2007).
  2. Almaca, J., Weitz, J., Rodriguez-Diaz, R., Pereira, E., Caicedo, A. The pericyte of the pancreatic islet regulates capillary diameter and local blood flow. Cell Metab. 27 (3), 630-644 (2018).
  3. Lifson, N., Lassa, C. V., Dixit, P. K. Relation between blood flow and morphology in islet organ of rat pancreas. Am J Physiol. 249 (1 Pt 1), E43-E48 (1985).
  4. Burganova, G., Bridges, C., Thorn, P., Landsman, L. The role of vascular cells in pancreatic beta-cell function. Front Endocrinol (Lausanne). 12, 667170 (2021).
  5. Hadi, H. A., Suwaidi, J. A. Endothelial dysfunction in diabetes mellitus. Vasc Health Risk Manag. 3 (6), 853-876 (2007).
  6. Granlund, L., Hedin, A., Korsgren, O., Skog, O., Lundberg, M. Altered microvasculature in pancreatic islets from subjects with type 1 diabetes. PLoS One. 17 (10), e0276942 (2022).
  7. Dai, C., et al. Pancreatic islet vasculature adapts to insulin resistance through dilation and not angiogenesis. Diabetes. 62 (12), 4144-4153 (2013).
  8. Nishikawa, Y., Tsuji, Y., Isoda, H., Kodama, Y., Chiba, T. Perfusion in the tissue surrounding pancreatic cancer and the patient’s prognosis. Biomed Res Int. 2014, 648021 (2014).
  9. Conchinha, N. V., et al. Protocols for endothelial cell isolation from mouse tissues: Brain, choroid, lung, and muscle. STAR Protoc. 2 (3), 100508 (2021).
  10. Ruck, T., Bittner, S., Epping, L., Herrmann, A. M., Meuth, S. G. Isolation of primary murine brain microvascular endothelial cells. J Vis Exp. (93), e52204 (2014).
  11. Wang, J., Niu, N., Xu, S., Jin, Z. G. A simple protocol for isolating mouse lung endothelial cells. Sci Rep. 9 (1), 1458 (2019).
  12. Sokol, L., et al. Protocols for endothelial cell isolation from mouse tissues: Small intestine, colon, heart, and liver. STAR Protoc. 2 (2), 100489 (2021).
  13. Guo, Q., Furuta, K., Aly, A., Ibrahim, S. H. Isolation and characterization of mouse primary liver sinusoidal endothelial cells. J Vis Exp. (178), e63062 (2021).
  14. Ieronimakis, N., Balasundaram, G., Reyes, M. Direct isolation, culture and transplant of mouse skeletal muscle derived endothelial cells with angiogenic potential. PLoS One. 3 (3), e0001753 (2008).
  15. Tang, X., et al. Suppression of endothelial ago1 promotes adipose tissue browning and improves metabolic dysfunction. Circulation. 142 (4), 365-379 (2020).
  16. Navone, S. E., et al. Isolation and expansion of human and mouse brain microvascular endothelial cells. Nat Protoc. 8 (9), 1680-1693 (2013).
  17. Chaurasiya, V., et al. Human visceral adipose tissue microvascular endothelial cell isolation and establishment of co-culture with white adipocytes to analyze cell-cell communication. Exp Cell Res. 433 (2), 113819 (2023).
  18. Villaret, A., et al. Adipose tissue endothelial cells from obese human subjects: Differences among depots in angiogenic, metabolic, and inflammatory gene expression and cellular senescence. Diabetes. 59 (11), 2755-2763 (2010).
  19. Domer, P., et al. Rapid and efficient immunomagnetic isolation of endothelial cells from human peripheral nerves. Sci Rep. 11 (1), 1951 (2021).
  20. Comhair, S. A., et al. Human primary lung endothelial cells in culture. Am J Respir Cell Mol Biol. 46 (6), 723-730 (2012).
  21. Hewett, P. W., Murray, J. C. Human lung microvessel endothelial cells: Isolation, culture, and characterization. Microvasc Res. 46 (1), 89-102 (1993).
  22. Gaskill, C., Majka, S. M. A high-yield isolation and enrichment strategy for human lung microvascular endothelial cells. Pulm Circ. 7 (1), 108-116 (2017).
  23. Malhi, N. K., et al. Isolation and profiling of human primary mesenteric arterial endothelial cells at the transcriptome level. J Vis Exp. (181), 63307 (2022).
  24. Reichard, A., Asosingh, K. Best practices for preparing a single cell suspension from solid tissues for flow cytometry. Cytometry A. 95 (2), 219-226 (2019).
  25. Brandhorst, H., et al. Large-scale comparison of liberase hi and collagenase nb1 utilized for human islet isolation. Cell Transplant. 19 (1), 3-8 (2010).
  26. Waise, S., et al. An optimised tissue disaggregation and data processing pipeline for characterising fibroblast phenotypes using single-cell rna sequencing. Sci Rep. 9 (1), 9580 (2019).
  27. Amsterdam, A., Jamieson, J. D. Studies on dispersed pancreatic exocrine cells. I. Dissociation technique and morphologic characteristics of separated cells. J Cell Biol. 63 (3), 1037-1056 (1974).
  28. Li, D., et al. Complete disassociation of adult pancreas into viable single cells through cold trypsin-edta digestion. J Zhejiang Univ Sci B. 14 (7), 596-603 (2013).
  29. Ji, B., Gaiser, S., Chen, X., Ernst, S. A., Logsdon, C. D. Intracellular trypsin induces pancreatic acinar cell death but not nf-kappab activation. J Biol Chem. 284 (26), 17488-17498 (2009).
  30. Mao, X., et al. Single-cell transcriptomic analysis of the mouse pancreas: Characteristic features of pancreatic ductal cells in chronic pancreatitis. Genes (Basel). 13 (6), 1015 (2022).
  31. Lee, H., Engin, F. Preparing highly viable single-cell suspensions from mouse pancreatic islets for single-cell rna sequencing. STAR Protoc. 1 (3), 100144 (2020).
  32. Schlesinger, Y., et al. Single-cell transcriptomes of pancreatic preinvasive lesions and cancer reveal acinar metaplastic cells’ heterogeneity. Nat Commun. 11 (1), 4516 (2020).
  33. Park, S., Dimaio, T. A., Scheef, E. A., Sorenson, C. M., Sheibani, N. Pecam-1 regulates proangiogenic properties of endothelial cells through modulation of cell-cell and cell-matrix interactions. Am J Physiol Cell Physiol. 299 (6), C1468-C1484 (2010).
  34. Kim, Y. W., et al. Integration of single-cell transcriptomes and biological function reveals distinct behavioral patterns in bone marrow endothelium. Nat Commun. 13 (1), 7235 (2022).
  35. Li, D. S., Yuan, Y. H., Tu, H. J., Liang, Q. L., Dai, L. J. A protocol for islet isolation from mouse pancreas. Nat Protoc. 4 (11), 1649-1652 (2009).
  36. Malhi, N. K., et al. Serine-arginine-rich protein kinase-1 inhibition for the treatment of diabetic retinopathy. Am J Physiol Heart Circ Physiol. 322 (6), H1014-H1027 (2022).
  37. Sachs, S., et al. Targeted pharmacological therapy restores beta-cell function for diabetes remission. Nat Metab. 2 (2), 192-209 (2020).
  38. Seaman, S. A., Tannan, S. C., Cao, Y., Peirce, S. M., Lin, K. Y. Differential effects of processing time and duration of collagenase digestion on human and murine fat grafts. Plast Reconstr Surg. 136 (2), 189e-199e (2015).
  39. Arbiser, J. L., et al. Oncogenic h-ras stimulates tumor angiogenesis by two distinct pathways. Proc Natl Acad Sci U S A. 94 (3), 861-866 (1997).
  40. Muraro, M. J., et al. A single-cell transcriptome atlas of the human pancreas. Cell Syst. 3 (4), 385-394 (2016).
  41. Segerstolpe, A., et al. Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes. Cell Metab. 24 (4), 593-607 (2016).
  42. Kang, R. B., et al. Single-nucleus rna sequencing of human pancreatic islets identifies novel gene sets and distinguishes beta-cell subpopulations with dynamic transcriptome profiles. Genome Med. 15 (1), 30 (2023).
  43. Lawlor, N., et al. Single-cell transcriptomes identify human islet cell signatures and reveal cell-type-specific expression changes in type 2 diabetes. Genome Res. 27 (2), 208-222 (2017).
  44. Fasolino, M., et al. Single-cell multi-omics analysis of human pancreatic islets reveals novel cellular states in type 1 diabetes. Nat Metab. 4 (2), 284-299 (2022).
  45. Yosefov-Levi, O., Tornovsky, S., Parnas, O. Pancreatic tissue dissection to isolate viable single cells. J Vis Exp. (195), e64871 (2023).
  46. Castillo, J. J., et al. Islet amyloid polypeptide aggregation exerts cytotoxic and proinflammatory effects on the islet vasculature in mice. Diabetologia. 65 (10), 1687-1700 (2022).
  47. Tremblay, J. R., et al. Rare, tightly-bound, multi-cellular clusters in the pancreatic ducts of adult mice function like progenitor cells and survive and proliferate after acinar cell injury. Stem Cells. 42 (4), 385-401 (2024).
  48. Zook, H. N., et al. Activation of ductal progenitor-like cells from adult human pancreas requires extracellular matrix protein signaling. iScience. 27 (3), 109237 (2024).
  49. Sordi, V., et al. Establishment, characterization and long-term culture of human endocrine pancreas-derived microvascular endothelial cells. Cytotherapy. 19 (1), 141-152 (2017).
This article has been published
Video Coming Soon
Keep me updated:

.

Diesen Artikel zitieren
Tapia, A., Kaur Malhi, N., Liu, X., Chen, M., Chen, Z. B. Isolation of Mouse Pancreatic Endothelial Cells. J. Vis. Exp. (208), e66690, doi:10.3791/66690 (2024).

View Video